Language selection

Search

Patent 3012277 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 3012277
(54) English Title: PREVENTION OF GRAFT REJECTION BY PRIOR USE OF MODIFIED GRAFTS
(54) French Title: PREVENTION DU REJET DE GREFFE PAR UTILISATION PREALABLE DE GREFFONS MODIFIES
Status: Report sent
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 35/12 (2015.01)
  • A61K 35/15 (2015.01)
  • A61K 35/17 (2015.01)
  • A61K 35/51 (2015.01)
  • A01N 1/02 (2006.01)
  • A61K 35/28 (2015.01)
  • A61K 39/395 (2006.01)
  • C07K 16/28 (2006.01)
(72) Inventors :
  • EMMRICH, FRANK (Germany)
  • FRICKE, STEPHAN (Germany)
  • HILGER, NADJA (Germany)
(73) Owners :
  • FRAUNHOFER-GESELLSCHAFT ZUR FOERDERUNG DER ANGEWANDTEN FORSCHUNG E.V. (Germany)
(71) Applicants :
  • FRAUNHOFER-GESELLSCHAFT ZUR FOERDERUNG DER ANGEWANDTEN FORSCHUNG E.V. (Germany)
(74) Agent: BENOIT & COTE INC.
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2017-02-15
(87) Open to Public Inspection: 2017-08-24
Examination requested: 2021-10-28
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/EP2017/053417
(87) International Publication Number: WO2017/140735
(85) National Entry: 2018-07-23

(30) Application Priority Data:
Application No. Country/Territory Date
16000373.7 European Patent Office (EPO) 2016-02-15

Abstracts

English Abstract

The present invention relates to an unmodified graft, a modified graft and/or a CD4 antagonist (preferably a CD4 antibody) for use in a method of treating one or more disease(s) treatable by transplantation in a subject, wherein said method comprises a first step of introducing into said subject a modified graft, and a second step of introducing into said subject said unmodified graft, wherein said modified graft is a cell graft containing immune cells, wherein said modification impairs CD4. The present invention also relates to various further uses of an unmodified graft, a modified graft and/or a CD4 antagonist.


French Abstract

La présente invention concerne un greffon non modifié, un greffon modifié et/ou un antagoniste du CD4 (de préférence un anticorps contre le CD4) destinés à être utilisés dans une méthode de traitement d'une ou de plusieurs maladie(s) traitables au moyen d'une greffe chez le patient, ladite méthode comprenant une première étape consistant à introduire dans le patient un greffon modifié, et une seconde étape consistant à introduire dans ledit patient ledit greffon non modifié, ledit greffon modifié étant un greffon cellulaire contenant des cellules immunitaires, ladite modification altérant le CD4. La présente invention concerne également diverses autres utilisations d'un greffon non modifié, d'un greffon modifié et/ou d'un antagoniste du CD4.

Claims

Note: Claims are shown in the official language in which they were submitted.


48
Claims
1. An unmodified graft for use in a method of treating one or more
disease(s) treatable
by transplantation in a subject,
wherein said method comprises a first step of introducing into said subject a
modified graft, and a second step of introducing into said subject said
unmodified
graft,
wherein said modified graft is a cell graft containing immune cells, wherein
said
modification inhibits CD4.
2. A modified graft for use in a method of treating one or more disease(s)
treatable by
transplantation,
wherein said method comprises a first step of introducing into said subject
said
modified graft, and a second step of introducing into said subject an
unmodified
graft,
wherein said modified graft is a cell graft containing immune cells, wherein
said
modification inhibits CD4.
3. A CD4 antagonist, preferably an anti CD4 antibody, for use in a method
of treating
one or more disease(s) treatable by transplantation;
wherein said method comprises a first step of introducing into said subject a
modified graft, and a second step of introducing into said subject an
unmodified
graft,
wherein said modified graft is a cell graft containing immune cells, wherein
said
modification inhibits CD4.
4. An unmodified graft, a modified graft and/or a CD4 antagonist, wherein
said
antagonist preferably is an anti CD4 antibody, for use in
i) a chirurgical transplantation method,
ii) a method of inducing transplantation tolerance, wherein a modified graft
is used
for inducing transplantation tolerance towards an unmodified graft,
iii) a method of transferring one or more cell suspension(s), particularly
stem cell
containing cell suspension(s) from a donor to a subject,
iv) a method of transferring one or more tissue(s) from a donor to a subject,
v) a method of transferring one or more partial organ(s) from a donor to a
subject,

49
vi) a method of transferring one or more organ(s) from a donor to a subject,
vii) a method of transferring hematopoietic system reconstituting cells from a
donor
source into a subject, optionally followed by transferring one or more
organ(s),
from said donor source into said subject,
or
viii) a method for enhancing immune reconstitution in a subject, optionally
followed by transferring one or more organ(s) into said subject;
wherein said method comprises a first step of introducing into said subject a
modified graft, and a second step of introducing into said subject an
unmodified
graft,
wherein said modified graft is a cell graft containing immune cells, wherein
said
modification inhibits CD4.
5. The unmodified graft for use, modified graft for use, or CD4 antagonist
for use of
any of the preceding claims,
wherein said first step is carried out from 30 min to 50 years, particularly
from one
hour to two months before said second step.
or
wherein said first step is carried out at least one hour, particularly at
least three
weeks before said second step.
6. The unmodified graft for use, modified graft for use, or CD4 antagonist
for use of
any of the preceding claims, wherein said modified graft comprises stem cells.
7. The unmodified graft for use, modified graft for use, or CD4 antagonist
for use of
any of the preceding claims,
wherein each of said grafts is selected from the group consisting of a cell
suspension, a tissue and an organ;
particularly
i) wherein said modified graft is a cell suspension and said graft is a cell
suspension,
or
ii) wherein said modified graft is a cell suspension and said graft is a
tissue,
or
iii) wherein said modified graft is a cell suspension and said graft is an
organ.
8. The unmodified graft for use, modified graft for use, or CD4 antagonist
for use of
any of the preceding claims, wherein said modified graft is a cell graft
containing

50
immune cells that comprises a CD4 antagonist, particularly an anti CD4
antibody,
particularly bound to CD4 epitopes thereof;
particularly wherein said modified graft
I) is obtainable by an in vitro method comprising the step a) incubating a
cell
graft containing immune cells with a CD4 antagonist, particularly an anti CD4
antibody, and optionally comprising the step b) removing unbound antibody
from said graft,
and/or
II) is a cell graft containing immune cells that comprises a CD4 antagonist,
particularly an anti CD4 antibody, bound to from 40 % to 100 % of the CD4
epitopes thereof.
9. The unmodified graft for use, modified graft for use, or CD4 antagonist
for use of
claim 8;
wherein said incubating in step a) of said in vitro method is carried out for
from 1
minute to 7 days,
particularly wherein said incubating is carried out
i) for from 1 to 150 minutes, particularly for from 5 minutes to 150 minutes,
more particularly for from 10 minutes to 150 minutes, more particularly for
from 30 minutes to 150 minutes, more particularly for from 40 minutes to
120 minutes, more particularly for from 45 minutes to 90 minutes,
especially for from 50 minutes to 70 minutes;
or
ii) for from 150 minutes to 7 days, particularly for from 150 minutes to 5
days, more particularly from 150 minutes to 3 days, more particularly from
150 minutes to 1 day, especially for from 150 minutes to 8 hours;
and/or
wherein said incubating in step a) of said in vitro method is carried out with
an
antibody amount of from 0.1 µg/ml to 10 mg/ml.
10. The unmodified graft for use, modified graft for use, or CD4 antagonist
for use of
any of the preceding claims, wherein said method
i) implies tolerance or partial tolerance within said unmodified graft against
the
recipient's tissue;
and/or
ii) implies tolerance or partial tolerance within the recipient's tissue
against said
unmodified graft;
and/or

51
iii) implies a reduced likelihood of developing any one of the group
consisting of
GvHD, donor graft rejection, and organ rejection, upon transplantation of the
unmodified graft.
11. The unmodified graft for use, modified graft for use, or CD4 antagonist
for use of
any of the preceding claims,
i) wherein said modified graft is selected from the group consisting of a
cell
suspension containing T cells and monocytes/macrophages the cell
suspension comprising bone marrow cells, non-adherent bone marrow cells,
peripheral blood cells, and/or cord blood cells; a cell suspension comprising
lymphocytes, monocytes and/or macrophages; a stem-cell-containing tissue;
a stem-cell-containing organ; an immune cell containing tissue; and an
immune cell containing organ;
and/or
ii) wherein said modified graft is a cell suspension and wherein said
method
comprises administration of an amount of from 2 x 10 6 cells to 2 x 10 15
nucleated cells to said subject.
12. The unmodified graft for use, modified graft for use, or CD4 antagonist
for use of
any of the preceding claims, wherein
A) the modified graft and the unmodified graft are from HLA-related donors;
particularly
i) wherein the said HLA-related donors comprise no more than a total of 50%

of HLA mismatches, preferably not more than 40% HLA mismatches,
preferably not more than 30% HLA mismatches, preferably not more than
20% HLA mismatches, preferably not more than 10% HLA mismatches,
preferably no HLA mismatch;
and/or
ii) wherein said HLA-related donors are selected from the group consisting of
tissue-type matched donors, related twins or are the same donor;
especially wherein both the modified graft and the unmodified graft are from
the
same donor,
and/or
B) wherein said modified graft and the unmodified graft are HLA-related;

52
particularly wherein said HLA-related grafts comprise no more than a total of
50% of HLA mismatches, preferably not more than 40% HLA mismatches,
preferably not more than 30% HLA mismatches, preferably not more than 20%
HLA mismatches, preferably not more than 10% HLA mismatches, preferably
no HLA mismatch.
13. The unmodified graft for use, modified graft for use, or CD4 antagonist
for use of
any of claims 3 to 12, wherein said CD4 antagonist is an anti CD4 antibody;
particularly wherein said anti CD4 antibody
i) is selected from the group consisting of Max 16H5, OKT4A, OKTcdr4a,

cMT-412, YHB.46, particularly wherein said anti CD4 antibody is
Max16H5;
or
11) is antibody 30F16H5;
or
iii) is obtainable from a cell line deposited with accession number
ECACC
88050502;
or
iv) is obtainable from a cell line MAX.16H5/30F16H5 deposited with the
DSMZ on December 2, 2011;
or
v) is antibody 16H5.chimIgG4;
or
vi) is obtainable from a cell line CD4.16H5.chimIgG4 deposited with the
DSMZ on December 2, 2011;
or
vii) is an antibody comprising the VH and the VK of antibody 16H5.chimIgG4;
or
viii) is an antibody comprising a VH and a VK of an antibody obtainable from a
cell line CD4.16H5.chimIgG4 deposited with the DSMZ on December 2,
2011;
or
ix) is an antibody comprising any combination of a VH selected from SEQ
ID
NOs: 1-10 and of a VK selected from SEQ ID NOs: 11-20, particularly
wherein said combination is selected from VH1/VK1 , VH2/VK2, VH4/VK2
and VH4/VK4, especially wherein said combination is VH2/VK2,
or

53
x) is a mixture of antibodies selected from the antibodies according
to i) to ix)
above.
14. The unmodified graft for use, modified graft for use, or CD4 antagonist
for use of
any of the preceding claims,
wherein, prior to introduction into said subject, said unmodified graft and/or

modified graft is/are additionally incubated with soluble bioactive molecules,

particularly with agents promoting immunosuppression, immunotolerance and/or
formation of regulatory T cells,
especially with an agent selected from the group consisting of 11-2, TGF-
.beta.,
rapamycin, retinoic acid, 4-1BB ligand, and anti CD28 antibodies; or any
combination thereof.
15. The unmodified graft for use, modified graft for use, or CD4 antagonist
for use of
any of the preceding claims,
A) wherein the modified graft is used
i) for achieving tolerance or partial tolerance within the modified graft
against
the recipient's tissue;
and/or
ii) for achieving tolerance or partial tolerance within the recipient's tissue
against the modified graft;
and/or
iii) for reducing the likelihood of any one of the group consisting of GvHD,
donor graft rejection, and organ rejection, upon transplantation of said
modified
graft,
and/or
B) wherein the modified graft is used
i) for achieving tolerance or partial tolerance within the unmodified graft
against
the recipient's tissue;
and/or
ii) for achieving tolerance or partial tolerance within the recipient's tissue
against the unmodified graft;
and/or
iii) for reducing the likelihood of any one of the group consisting of GvHD,
donor graft rejection, and organ rejection, upon transplantation of said
unmodified graft.

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 03012277 2018-07-23
WO 2017/140735 - 1 - PCT/EP2017/053417
Prevention of graft rejection by prior use of modified grafts
The invention generally relates to the field of grafts and transplantations
thereof. In
particular, the invention relates to modified grafts and unmodified grafts for
use in methods
of treating diseases treatable by transplantation as well as related uses. The
said methods
generally comprise a step of introducing into a subject a modified graft, and
a second step
of introducing into said subject an unmodified graft. The invention makes use
of inhibition
of CD4 e.g. in said modified grafts.
In certain aspects, the invention relates to the field of solid organ grafts
and
transplantations thereof The invention relates to modified grafts, where
introduction
thereof is e.g. followed by transplantation of a solid organ. According to the
invention the
need for conventionally immunosuppressive drugs may be avoided. The invention
also
describes methods of obtaining the modified grafts to be used in the
invention. In certain
aspects, the invention relates to modification of allogeneic grafts containing
immune
competent viable cells by anti human CD4 antibodies before transplantation of
solid
organs.
BACKGROUND OF THE INVENTION
In many cases, transplantation of solid organs is still the only curative
treatment for many
patients with organ dysfunction. Transplantations of solid organs are well
established
methods in medicine and were performed for circa 50 years [Sayegh, 2004]. The
investigation of the blood types A, B, 0 was done by K. Landsteiner in 1901
[Landsteiner,
1901; Durand & Willis, 2010] followed by AB by Alfredo von Castello and
Adriano Sturli
[de Castello & Sturli, 1902]. The first transplantation of a kidney in animals
was performed
by Alexis Carrel (1873-1944) in 1906 [Carrel & Guthrie, 1906]. A milestone in
the
development of solid organ transplantation was the investigation of the tissue
compatibility
and immune reactions during skin transplantation by Sir Peter Medawar (1915-
1987) in
1944 [Medawar, 1944]. The first syngeneic and allogeneic human kidney
transplantations
were achieved by Joseph E. Murray in 1954 and 1959, respectively [Murray et
al., 1976],
and the HLA antigens responsible for graft rejection were detected by Jean
Dausset in
1958 [Dausset, 1958]. In 1962, the use of the immunosuppressive drug
azathioprine
permitted the kidney transplantation of a deceased person [Calne et al.,
1962]. In Germany,

CA 03012277 2018-07-23
WO 2017/140735 - 2 - PCT/EP2017/053417
the first (clinical) kidney transplantation between relatives was done by
Wilhelm Brosig in
1963 [Eigler, 2002]. First transplantation of pancreas was done by Richard
Lillehei in 1966
[Kelly et al., 1967], first successful transplantation of a liver by Tom
Starzl in 1967 [Starzl
et al., 1968; Starzl et al., 1982]. Also in 1967, the first heart
transplantation was performed
by Christiaan N. Barnard (1922-2001) in Cape Town, South Africa [Barnard et
al., 1967],
followed by the first transplantation of heart and lung by Denton A. Cooley in
1969
[Cooley et al., 1969]. For protection of graft rejection, cyclosporin A was
first used in 1983
[Kapturczak, 2004]. From the first organ transplantation in 1963 until 2013,
116.650 solid
organ transplantations were conducted in Germany [Eurotransplant, 2015a].
in .. In January 2015, 12.500 solid organs were needed, which is more than
available
[Eurotransplant, 2015b]. Moreover, allogeneic solid organ transplantation can
be
associated with severe complications. Graft rejection due to genetic
disparities, side
effects, toxicity, infections and the development of secondary tumors due to
life-long
treatment with immunosuppressive drugs are serious side effects [Hsu &
Katelaris, 2009].
Also the mental impact for the patients due to waiting for a (matching) solid
organ graft
should be considered [Schulz & Koch, 2005].
Strategies for prophylaxis of graft rejection by conventional
immunosuppressive drugs
have not been able to distinguish between different T cell clones and
therefore not between
T cell clones responsible for graft rejection and responsible for maintenance
of healthy
.. immunological reactions (e.g., destruction of bacteria, viruses, tumor
cells). Generally, the
use of conventionally immunosuppressive drugs can lead to suppression of the
entire
immune system, which enhances the possibility for infections and/or
development of
malignant tumors [Fricke et al. 2012, 2014].
The investigation of alternative or improved therapeutic approaches or
procedures for
.. prevention of graft rejection after solid organ transplantation without
conventional
immunosuppressive drugs by not ameliorating the desired healthy immunological
reactions
is still needed.
In a previous work depicted in patent application WO 2012/072268, which is
incorporated
by reference herein in its entirety, it was e.g. shown that an in vitro method
of modifying a
graft comprising the steps of a) incubating a graft with an anti-CD4 antibody
wherein said
incubating is carried out for from 1 min to 7 days and b) removing unbound
antibody from
said graft, allowed the prevention of Graft-versus-Host-Disease (GvHD) and not

decreasing the Graft-versus-Tumor-effect in allogeneic hematopoietic stem cell

transplantation at the same time due to induction of immune tolerance. This
immune
tolerance is considered to be due to modulation of CD4 ' T cells by MAX.16H5
IgGi or
CD4.16H5.chimIgG4. CD4 molecules directly bind to constant regions of HLA
molecules

CA 03012277 2018-07-23
WO 2017/140735 - 3 - PCT/EP2017/053417
of antigen presenting cells (APCs) to allow complete T cell activation [In:
Murphy KM,
Travers P, Walport M., English translation by Seidler L, Hau13er-Siller I.
(Hrsg):
Immunologie, 7. Edition -Heidelberg, Berlin: Spektrum, Akad. Verl. 2008]. To
interfere
with this binding by non-depleting monoclonal antibodies may inhibit this
activation by a
total steric blockage, by shortening of cell-cell contact between APC and T
cell [cf.
Harding, 2002], by induction of negative signals through inhibition of protein
tyrosine
phosphorylation [June et al., 1990] or induction of T cell anergy [cf.
Madrenas, 1996a;
Madrenas, 1996b].
CD4 is a surface glycoprotein primarily expressed on cells of the T lymphocyte
lineage
including a majority of thymocytes and a subset of peripheral T cells
[Harrison, 1993;
Luckheeram et al., 2012]. Low levels of CD4 are also expressed by some non-
lymphoid
cells [Stewart et al., 1986]. On mature T cells, CD4 functions as a co-
recognition signal
through interaction with MHC Class II molecules expressed on antigen
presenting cells
[Harrison, 1993]. CD4 T cells primarily constitute the helper subset which
regulates T and
B cell functions during T-dependent responses to e.g. viral, bacterial, fungal
and parasitic
infections [Jiang & Dong, 2013]. During the pathogenesis of autoimmune
diseases, in
particular when tolerance to self-antigens breaks down, CD4 ' T cells
contribute to
inflammatory responses which inter alia result in tissue destruction [Bellone,
2005]. These
processes are facilitated by the recruitment of inflammatory cells of the
hematopoietic
lineage, production of antibodies, inflammatory cytokines and mediators, and
by the
activation of killer cells [Bellone, 2005].
Previously, the murine anti human CD4 monoclonal antibody MAX.16H5 IgGi was
used
in patients with autoimmune diseases or as a protective therapy against
transplant rejection
[Emmrich, 1991a; Emmrich, 1991b; Reinke, 1991]. In human kidney
transplantation,
MAX.16H5 IgGi had the potential to effectively reduce graft rejection [Reinke,
1991;
Reinke, 1994; Reinke, 1995]. The use of MAX.16H5 IgGi not only resulted in
suppression
of immune activity but also in the induction of tolerance against tetanus
toxoid in a triple
transgenic mouse model [Laub, 2000; Laub, 2001; Laub, 2002; Strauss, 1994;
Fricke,
2014]. The development of these transgenic mice was performed by scientists
mentioned
in the literature [Laub, 2000; Laub, 2001; Laub, 2002; Strauss, 1994]. CD4/DR3
mice
expressing human CD4 and HLA-DR17, a split antigen of HLA-DR3, on a murine CD4-

deficient background, were initially bred at the Institute for Laboratory
Animal Science,
Medical School Hanover (Germany) [Fricke, 2014]. Using these mice, anti human
CD4
antibodies can be used directly, and host and donor hematopoiesis could be
distinguished
by means of human and murine CD4 molecules [Fricke, 2014; Cooke, 1998;
O'Connell,
2010; Liu, 2006]. In a T cell activating environment, MAX.16H5 IgGi
preincubated CD4 '
T cells were shown to express lower IL-2 mRNA levels and not to activate Lck
dependent

CA 03012277 2018-07-23
WO 2017/140735 - 4 - PCT/EP2017/053417
signal transduction in contrast to controls without prior MAX.16H5 IgGi
incubation
[Fricke, 2014].
Direct application of antibodies to human beings can be associated with an
unwanted
immune reaction to the therapeutic protein [de Groot & Scott, 2007]. Several
mouse
monoclonal antibodies have shown promise as therapies in a number of human
disease
settings but in certain cases have failed due to the induction of significant
degrees of a
human anti-murine antibody (HAMA) response [Becker, 2012]. The direct
application of
antibodies to patients may results in severe anaphylactic reactions [de Groot
& Scott,
2007].
In previous work, a murine GvHD and tumor transplantation model was shown that
allows
the direct testing of anti human CD4 antibodies for long-term prevention of
GvHD [Fricke,
2014]. However, advantageously, the GyL ("Graft-versus-Leukemia") effect was
not
decreased [Fricke, 2014]. Without intending to be bound by theory, it may be
considered
that this effect is CD4 epitope specific [Fricke, 2014]. Advantageously, GvHD
was
prevented when the graft was short-term pre-incubated (2 hours) with MAX.16H5
IgGi
and unbound antibodies were removed [Fricke, 2014]. A direct application of
the
antibodies was not necessary [Fricke, 2014].
However, there still remains a need for methods for introduction of grafts,
particularly of
non-modified grafts. Also, there still remains a need for advantageous ways of
treating
diseases treatable by transplantation of grafts. Generally, there still
remains a need for
promising alternative or improved therapeutic approaches that lack
disadvantages of the
prior art methodologies.
SUMMARY OF THE INVENTION
In one aspect, the present invention relates to an unmodified graft for use in
a method of
treating one or more disease(s) treatable by transplantation in a subject,
wherein said
method comprises a first step of introducing into said subject a modified
graft, and a
second step of introducing into said subject said unmodified graft, wherein
said modified
graft is a cell graft containing immune cells, wherein said modification
inhibits CD4.
In a second aspect, the present invention relates to a modified graft for use
in a method of
.. treating one or more disease(s) treatable by transplantation, wherein said
method comprises
a first step of introducing said modified graft into said subject, and a
second step of
introducing an unmodified graft into said subject, wherein said modified graft
is a cell graft
containing immune cells, wherein said modification inhibits CD4.

CA 03012277 2018-07-23
WO 2017/140735 PCT/EP2017/053417
- 5 -
In a third aspect, the present invention relates to a CD4 antagonist for use
in a method of
treating one or more disease(s) treatable by transplantation; wherein said
method
comprises a first step of introducing into said subject a modified graft, and
a second step of
introducing into said subject an unmodified graft, wherein said modified graft
is a cell graft
containing immune cells, wherein said modification inhibits CD4.
In a fourth aspect, the present invention relates to an unmodified graft, a
modified graft
and/or a CD4 antagonist for use in i) a chirurgical transplantation method,
ii) a method of
inducing transplantation tolerance, wherein a modified graft is used for
inducing
transplantation tolerance towards an unmodified graft, iii) a method of
transferring one or
more cell suspension(s), particularly stem cell containing cell suspension(s)
from a donor
to a subject, iv) a method of transferring one or more tissue(s) from a donor
to a subject, v)
a method of transferring one or more partial organ(s) from a donor to a
subject, vi) a
method of transferring one or more organ(s) from a donor to a subject, vii) a
method of
transferring hematopoietic system reconstituting cells from a donor source
into a subject,
.. optionally followed by transferring one or more organ(s) from said donor
source into said
subject, and/or viii) a method for enhancing immune reconstitution in a
subject, optionally
followed by transferring one or more organ(s) into said subject; wherein said
method
comprises a first step of introducing into said subject a modified graft, and
a second step of
introducing into said subject an unmodified graft, wherein said modified graft
is a cell graft
containing immune cells, wherein said modification inhibits CD4.
Generally herein, in preferred embodiments, the CD4 antagonist is a CD4
antibody.
Generally herein, in preferred embodiments, the modified graft and the
unmodified graft
are from HLA-related donors.
SHORT DESCRIPTION OF THE FIGURES
.. Fig. 1 shows Balb/c (TTG) mice and wildtype Balb/c' t (control) mice 24h
and 50 days (d)
after transplantation of skin from TTG donor mice. No graft rejection could be
observed in
Balb/c (TTG) mice.
Fig. 2 contains an explanation of C57B1/6-triple transgenic mice (TTG) as
donors. TTG
mice express human CD4 and HLA-DR while murine CD4 molecules are knocked out.
That allows the determination of specific human surface molecules after
transplantation
and the direct testing of anti-human CD4 antibodies in mice.
Fig. 3 shows the explanation of an experimental design herein. Bone marrow
cells and
splenocytes were taken from TTG mice, pooled, incubated in medium containing
either
anti-human CD4 antibodies or no antibodies for 2 hours, and transplanted in
lethally

CA 03012277 2018-07-23
WO 2017/140735 - 6 - PCT/EP2017/053417
irradiated Balb/cwt mice. The experiment results were compared with results
obtained by
using donor cells from C57B1/6 wild-type mice. Therapeutic effects after
transplantation
were investigated.
Moreover, SEQ ID NOs 1-10 depict the DNA and amino acid sequences of exemplary
modified heavy chain variable regions of a particular anti-CD4 antibody for
use in the
invention.
SEQ ID NOs 11-20 depict the DNA and amino acid sequences of exemplary modified
light
chain variable regions of a particular anti-CD4 antibody for use in the
invention.
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
HI The present invention solves above described objects and overcomes above
described
deficiencies of prior art methods. It includes the first report showing that
ex vivo
modulation of an allogeneic hematopoietic stem cell graft by anti-human CD4
antibodies
MAX.16H5 IgGi followed by third party skin transplantation prevents skin graft
rejection
in a full MHC-mismatch transplantation model.
Without intending to be bound by theory, the present inventors consider that
differentiation
of or distinction between T cell clones responsible for graft rejection and
those responsible
for maintenance of healthy immunological reactions could be achieved by
modulation of
CD4 T-helper cells.
Surprisingly, and further to the previous work described herein above, the
present
inventors e.g. found that GvHD was not re-induced after single incubation of
an allogeneic
graft by MAX.16H5 IgGi even when cells were transferred in a third party
recipient
without re-incubation of MAX.16H5 IgGi. Also, GvL was not prevented. To the
inventor's
knowledge, such a therapy for induction of tolerance with regard to GvHD by
preserving
the GvL effect was surprising. Without intending to be bound by theory, the
present
inventors consider that, advantageously, the maintenance of the immunological
effect (e.g.
freedom from GvHD) was induced at the day of transplantation and kept up by
regulatory
T cells.
Among others, the present inventors provide the first report showing that ex
vivo
modulation of an allogeneic hematopoietic stem cell graft by anti human CD4
antibodies
MAX.16H5 IgGi followed by third party skin transplantation prevents skin graft
rejection
in a full MHC-mismatch transplantation model.
Currently used drugs for prevention of graft rejection are associated with
side effects and
do not distinguish between different T cell clones and therefore not between T
cell clones

CA 03012277 2018-07-23
WO 2017/140735 PCT/EP2017/053417
- 7 -
responsible for graft rejection and responsible for maintenance of healthy
immunological
reactions (e.g., destruction of bacteria, viruses, tumor cells) [Fricke,
2014].
Inter alia, anti-human CD4 antibodies MAX.16H5 IgGi were tested in murine stem
cell
transplantation and in a skin transplantation model. Ex vivo modified grafts
from human
CD4 ' C57B1/6 mice (cf. Fig. 2) were transplanted into Balb/cwt mice. After
119 days, third
party skins from the human CD4 ' C57B1/6 mice were transplanted to these
recipient mice.
The survival rate of these skin grafts was significantly increased in
recipients receiving a
MAX.16H5 IgGi short-term (2 hours) pre-incubated graft (cf. Fig. 1). A direct
application
of the antibodies to the recipients was not necessary. The anti-human CD4
antibodies
MAX.16H5 IgGi were long-term effective with regard to survival of solid skin
grafts. The
nature of the observed induction of immune tolerance by short-term pre-
incubation of an
antibody with regard to solid organs without the need of conventionally
immunosuppressive drugs was advantageous and surprising.
These findings are considered to e.g. make allogeneic organ transplantation
safer, and
prevent graft rejection due to genetic disparities, reduce side effects,
toxicity, and
infections as well as the development of secondary tumors due to life-long
treatment with
immunosuppressive drugs.
Within this patent application, the present inventors could e.g. show that a
prior use of a
modified graft is suitable to achieve tolerance to an unmodified graft, e.g.
in solid organ
transplantation (cf. Fig. 3).
Accordingly, the methods of the present invention are e.g. useful i) for
reducing the
likelihood of donor graft rejection, and organ rejection of a third or higher
party organ; ii)
for achieving tolerance within the transplanted immunocompetent cells against
the third or
higher party organ; iii) for achieving tolerance or partial tolerance within
the recipient's
tissue against the third or higher party organ; and/or iv) for silencing cell
activation against
a third or higher party organ.
The observed induction of immune tolerance by short-term pre-incubation of an
antibody
with regard to solid organs without the need for conventional
immunosuppressive drugs is
advantageous. The present findings are considered to make e.g. allogeneic
organ
transplantation safer, prevent graft rejection due to genetic disparities, and
reduce side
effects, toxicity, and infections as well as the development of secondary
tumors because
life-long treatment with immunosuppressive drugs may be unnecessary.
In certain embodiments, the present invention uses the in vitro treatment of
cell grafts
containing immune cells with antibodies, thereby avoiding their direct
application in vivo.

CA 03012277 2018-07-23
WO 2017/140735 - 8 - PCT/EP2017/053417
In detail, the present invention may e.g. use a short term incubation of a
(stem cell) graft
such as cell suspensions containing T cells, in particular CD4 ' cells, with
the aim of
tolerance induction or immunosuppression. Without intending to be bound by
theory, it is
considered that e.g. the anti-CD4 antibody incubation of (stem cell) grafts
comprising CD4
positive (immune) cells and subsequent removing of unbound antibodies,
preferably
removing of only unbound antibodies, results in a modified graft, wherein the
antibody
labeled cells are are not activated, in particular not activated as soon as
they encounter
specific antigen. Preferably, the antibody labeled cells in the modified graft
are anergic
cells. Accordingly, e.g. GvHD is not initiated.
Without intending to be bound by theory, it is envisaged that the CD4
antagonist such as
anti-CD4 antibody may for example inhibit immune cells (such as lymphocytes)
bearing
CD4 (e.g. by binding to them) and thereby exerts its beneficial effect. Also,
as will be
readily apparent to the skilled person, substantially reducing or avoiding the
administration
of free CD4 antagonists (such as anti-CD4 antibodies, i.e. anti-CD4 antibodies
that are not
bound to an antigen located on the graft) is advantageous.
In any case, it is considered feasible that CD4 is antagonized by any other
way. Such
alternative ways of inhibiting CD4 are well-known and easily appliable by the
skilled
person. CD4 antagonists are well studied and numerous options are known and
available to
the skilled person ¨ and are e.g. further described herein below.
In general, practicing the present invention is considered to involve one or
more of the
following advantages: i) no direct application of the CD4 antagonist (such as
anti-CD4
antibodies) to the graft recipients is required; ii) suitable use of a short-
term incubation of
the graft, such as cell suspensions, tissues, and organs containing T cells,
in particular
CD4 ' cells; iii) GvHD prevention; iv) prevention of other immunological
complications
after transplantation of the graft of the invention (e.g. cytokine-release
syndrome); v)
reduction of costs due to the avoided treatment with conventional
immunosuppressive
drugs or reduction of their dosage and the significantly reduced amount of
antibodies as
compared to systemic application; vi) improvement of survival of patients
receiving (a)
graft(s) in accordance with the invention; vii) facilitation of
transplantation of grafts also
into patients, into whom regular grafts cannot be transplanted due to expected

immunological complications, such as older patients; viii) transplantation of
grafts from
HLA mismatch donors or grafts from less good HLA matched donors than without
the
invention; ix) maintenance of the GvL effect; and/or x) facilitation of
repeated
transplantation of grafts into a subject.
Generally, the present invention relates to subject matter as defined in the
claims and
herein below.

CA 03012277 2018-07-23
WO 2017/140735 - 9 - PCT/EP2017/053417
That is, in a first aspect, the present invention relates to an unmodified
graft for use in a
method of treating one or more disease(s) treatable by transplantation in a
subject, wherein
said method comprises a first step of introducing into said subject a modified
graft, and a
second step of introducing into said subject said unmodified graft, wherein
said modified
graft is a cell graft containing immune cells, wherein said modification
inhibits CD4 (or
involves CD4 inhibition, respectively).
In a second aspect, the present invention relates to a modified graft for use
in a method of
treating one or more disease(s) treatable by transplantation, wherein said
method comprises
a first step of introducing into said subject said modified graft, and a
second step of
introducing into said subject an unmodified graft, wherein said modified graft
is a cell graft
containing immune cells, wherein said modification inhibits CD4 (or involves
CD4
inhibition, respectively).
In a third aspect, the present invention relates to an CD4 antagonist,
preferably an anti-
CD4 antibody, for use in a method of treating one or more disease(s) treatable
by
transplantation; wherein said method comprises a first step of introducing
into said subject
a modified graft, and a second step of introducing into said subject an
unmodified graft,
wherein said modified graft is a cell graft containing immune cells, wherein
said
modification inhibits CD4 (or involves CD4 inhibition, respectively).
Preferably, the said modification involves use of the said CD4 antagonist.
Preferably, the
said modification is effected by the said CD4 antagonist.
Generally herein, the subject may also be referred to as recipient. Preferably
herein, the
subject is an animal, especially a mammal, particularly a human.
Likewise, preferably herein, the donor(s) (from which the graft(s) is/are
isolated and/or
derived) is/are an animal, especially a mammal, particularly a human.
Generally herein, the
modified and unmodified graft(s) may be from the same donor or from different
donors.
Preferred embodiments with respect to the donor(s) are defined elsewhere
herein.
Preferably, said one or more diseases treatable by transplantation is/are
selected from the
group consisting of acute myeloid leukemia (AML); acute lymphoid leukemia
(ALL);
chronic myeloid leukemia (CML); myelodysplastic syndrome (MDS) /
myeloproliferative
syndrome; malign lymphomas, particularly selected from Morbus Hodgkin, high
grade
Non-Hodgkin Lymphoma (NHL), mantle cell lymphoma (MCL), low malign NHL,
chronic lymphatic leukemia (CLL), multiple myeloma; severe aplastic anemia;
thalassemia; sickle cell anemia; immunological defects particularly selected
from severe
combined immunodeficiency (SCID), Wiskott-Aldrich syndrome (WAS), and

CA 03012277 2018-07-23
WO 2017/140735 - 10 - PCT/EP2017/053417
hemophagocytic lymphohistiocytosis (HLH); inborn errors of metabolism
particularly
selected from lysosomal storage disorders and disorders of peroxisomal
function;
autoimmune diseases; rheumatologic diseases; and recidivisms of any of the
above.
Even more preferably, said one or more diseases are one or more hematological
malignancies especially selected from acute myeloid leukemia (AML); acute
lymphoid
leukemia (ALL); chronic myeloid leukemia (CML); myelodysplastic syndrome (MDS)
/
myeloproliferative syndrome; malign lymphomas, particularly selected from
Morbus
Hodgkin, high grade Non-Hodgkin lymphoma (NHL), mantle cell lymphoma (MCL),
low
malign Non-Hodgkin lymphoma (NHL), chronic lymphatic leukemia (CLL), multiple
myeloma; severe aplastic anemia; thalassemia; and sickle cell anemia.
In certain embodiments herein, the disease treatable by transplantation is any
condition
requiring transplantation.
In certain preferred embodiments herein, the disease treatable by
transplantation is a
disease involving organ malfunction. Accordingly, said disease may e.g.
require
transplantation to replace an organ, which does not function as desired.
In certain embodiments herein, the disease treatable by transplantation is
transplant
rejection, such as organ rejection. Further examples are GvHD and donor graft
rejection.
That is to say, in the latter cases, a previously transplanted transplant,
such as an organ,
may be replaced by another transplant, such as an organ, in line with the
present invention.
Generally herein, said one or more diseases treatable by transplantation also
include
recidivisms of any of the above as well as any combination of diseases
mentioned herein.
As used herein, a "cell graft containing immune cells" essentially refers to a
graft
comprising immune cells. Generally, the cell graft containing immune cells is
not
particularly limited, with particular embodiments corresponding to those
described herein
below, e.g. in context with the modified graft.
Generally, grafts, and also cell grafts containing immune cells, are very well
known to the
person skilled in the art. Also the use of grafts including cell grafts
containing immune
cells in transplantation is well known in the art. According to the present
invention, the
graft may comprise a cell suspension, a tissue and/or an organ. Preferably,
the graft is a
cell suspension, a tissue and/or an organ. More preferably, the grafts are
selected from the
group consisting of a cell suspension, a tissue and an organ. Generally,
herein, a graft may
also be a combination of grafts, such as a combination of one or more of the
grafts referred
to above, e.g. a combination of an organ and a cell suspension. In another
preferred
embodiment, the graft is not an artificial graft. In a preferred embodiment,
it is understood

CA 03012277 2018-07-23
WO 2017/140735 - 11 - PCT/EP2017/053417
that grafts, and also cell grafts containing immune cells, are intended for
and useful for
therapeutic purposes in vivo, in particular intended for and useful for
treating a disease
involving organ malfunction. Said disease may e.g. require transplantation to
replace an
organ, which does not function as desired. Accordingly, in another preferred
embodiment,
grafts, and also cell grafts containing immune cells, exclude cells and
compositions
comprising cells, which are intended for and/or useful only for mechanistic
studies in
animal models, which do not aim at a therapy of a disease.
In preferred embodiments herein, each of the said grafts is selected from the
group
consisting of a cell suspension, a tissue and an organ. Accordingly, in the
present
invention, the modified graft may be selected from the group consisting of a
cell
suspension, a tissue and an organ. Accordingly, in the present invention, the
unmodified
graft may be selected from the group consisting of a cell suspension, a tissue
and an organ.
In particular embodiments, said modified graft is a cell suspension and said
graft is a cell
suspension. In particular embodiments, said modified graft is a cell
suspension and said
graft is a tissue. In particular embodiments, said modified graft is a cell
suspension and
said graft is an organ.
In some particular embodiments the graft does not include embryonic stem cells
that are
not derived and/or reprogrammed from adult stem cells. Hence, in some
particular
embodiments any embryonic stem cells included in the graft are embryonic stem
cells that
are derived and/or reprogrammed from adult stem cells. In some particular
embodiments,
the grafts do not consist of or do not comprise totipotent embryonic stem
cells. In certain
embodiments herein, the grafts do not consist of or do not comprise embryonic
stem cells.
In particular embodiments herein, the grafts do not comprise embryonic stem
cells derived
from a human embryo.
As used herein, an unmodified graft is a regular graft as it is well-known to
a skilled
person. In particular, it will be readily understood by the skilled person
that such
unmodified graft is not modified as the modified graft described herein, i.e.
does not
comprise the modifications of the modified graft herein. Accordingly, the
unmodified graft
may be described as not having been modified by use of a CD4 antagonist.
Accordingly,
.. the unmodified graft may be described as not comprising CD4 antagonists
(such as anti-
CD4 antibodies). Accordingly, the unmodified graft may be described as not
comprising a
modification that inhibits CD4. Preferred particular embodiments of an
unmodified graft
are described herein below.
The present invention e.g. advantageously uses a modified graft. Generally,
modified
.. grafts used in the invention may be used as it is known in the art for
unmodified grafts, e.g.
as regards their introduction (e.g. transplantation) into a subject.

CA 03012277 2018-07-23
WO 2017/140735 - 12 - PCT/EP2017/053417
As used herein, a modified graft is a graft that has been modified (preferably
that has
artificially been modified), e.g. via a method described herein, preferably by
an in vitro
method described herein, preferably by use of a CD4 antagonist. Hence, said
modification
may be said to involve use of a CD4 antagonist. Likewise, said modification
may be said to
be effected by a CD4 antagonist. The modified graft herein may have been
modified prior
to the uses of the invention or during the uses of the invention.
Generally herein, the modification of the modified graft is said to inhibit
CD4. Hence, the
modified graft is preferably described as comprising a modification that
inhibits CD4.
Accordingly, it may be said that the modified graft herein has been / is
modified to inhibit
CD4.
Generally, inhibition as referred to herein may not mean absolute inhibition
but also
includes partial inhibition. Sufficient degrees of inhibition may readily be
determined by
the skilled person. Particular embodiments of degrees of inhibition e.g.
correspond to
embodiments (e.g. including the percentage values recited in context with
antibody binding
to a graft) of the modified graft described elsewhere herein. A particular
preferable degree
of preferred inhibition achieves at least one of the advantageous effects
disclosed herein.
As used herein, a "modified graft wherein the modification inhibits CD4" ("...
involves
CD4 inhibition", or other suchlike and/or equivalent terms) preferably refers
to a modified
graft, where the expression and/or function, preferably the function, of CD4
is inhibited (or
impaired). Ways to inhibit CD4 are known to the skilled person and are not
particularly
limited. They may involve use of (or be effected by, respectively) any CD4
antagonist(s),
and particularly of any antagonist(s) of CD4 described herein. Accordingly, in
any of the
aspects herein, a CD4 antagonist (or e.g. also a mixture of CD4 antagonists)
may be used.
In certain embodiments herein, as the skilled reader will readily appreciate,
the term
"wherein the modification inhibits CD4" and suchlike terms is/are replaced by
the term
"wherein the modification inhibits cellular functions via CD4". In a preferred
embodiment,
the term "wherein the modification inhibits CD4" and suchlike terms is/are
replaced by the
term "wherein the modification inhibits or impairs cellular functions
downstream of CD4,
in particular binding and/or recruitment". Cellular functions that may be
inhibited via CD4
are readily known to the skilled person, particularly in view of his knowledge
on CD4 and
its mode of action in the cell. In certain embodiments herein, at least one
such cellular
function is inhibited.
Likewise, in certain embodiments herein, the term "wherein the modification
involves
CD4 inhibition" and suchlike terms is/are replaced by the term "wherein the
modification
involves CD4-mediated inhibition of function". In one preferred embodiment,
the term

CA 03012277 2018-07-23
WO 2017/140735 - 13 - PCT/EP2017/053417
"wherein the modification involves CD4 inhibition" and suchlike terms is/are
replaced by
the term "wherein the modification involves inhibition or impairment of
function". In
another preferred embodiment, the term "wherein the modification involves CD4
inhibition" and suchlike terms is/are replaced by the term "wherein the
modification
involves inhibition or impairment of CD4-mediated function"
Generally, non-limiting embodiments of CD4 antagonists to be used in context
with the
present invention include CD4 antagonists, which modulate CD4 expression
and/or
function. Accordingly, in certain embodiments a CD4 antagonist herein may
modulate,
particularly inhibit, expression of CD4. Accordingly, in certain embodiments a
CD4
antagonist herein may modulate, particularly inhibit, the function of CD4 ¨
especially by
binding to it. Accordingly, the CD4 antagonists herein may be CD4 ligands.
Generally,
CD4 antagonists such as CD4 inhibitors or CD4 ligands, respectively, are known
in the art.
Numerous CD4 antagonists, and particularly CD4 ligands that bind to CD4, are
commercially available.
CD4 antagonists are generally well known in the art. Non-limiting examples
include the
ones described herein. Generally herein, said CD4 antagonists, e.g. said CD4
inhibitors or
CD4 ligands, respectively, may be extra- or intracellular agents (such as
ligands).
Generally herein, and particularly also in the third aspect, said CD4
inhibition may directly
or indirectly be caused by said CD4 antagonist.
In preferred embodiments herein, the CD4 antagonist binds to CD4.
In particularly preferred embodiments herein, the CD4 antagonist is an anti-
CD4 antibody.
Anti-CD4 antibodies are generally well-known in the art. Numerous anti-CD4
antibodies
are commercially available.
Antibodies and also anti-CD4 antibodies are well known in the art. As used
herein, by
"antibody" is meant inter alia a protein of the immunoglobulin family that is
capable of
specifically combining, interacting or otherwise associating with an antigen,
wherein said
combining, interacting or otherwise associating (such as binding) of the
antibody to the
antigen is mediated by complementarity-determining regions (CDRs). Similarly,
term
"antigen" is used herein to refer to a substance that is capable of
specifically combining,
interacting or otherwise associating with said antibody. In the context of the
anti-CD4
antibody of the present invention the antigen is meant to be CD4, particularly
human CD4.
As used herein, the term "CDR" refers to the "complementarity-determining
region" of an
antibody, i.e. to one of the hypervariable regions within an immunoglobulin
variable
domain contributing to the determination of antibody specificity. CDRs are
well known to
a person skilled in the art. Typically, both the heavy chain immunoglobulin
variable

CA 03012277 2018-07-23
WO 2017/140735 - 14 - PCT/EP2017/053417
domain and the light chain immunoglobulin variable domain contain three CDRs.
In the
context of the present invention, the term "antibody" is considered to also
relate to
antibody fragments including for example Fv, Fab, Fab' and F(ab')2 fragments.
Such
fragments may be prepared by standard methods [for example; Coligan et al.,
1991-1997,
incorporated herein by reference]. The present invention also contemplates the
various
recombinant forms of antibody derived molecular species well known in the art.
Such
species include stabilized Fv fragments including single chain Fv forms (e.g.,
scFv)
comprising a peptide linker joining the VH and VL domains, or an Fv stabilized
by
interchain disulphide linkage (dsFv) and which contain additional cysteine
residues
engineered to facilitate the conjoining of the VH and VL domains. Equally,
other
compositions are familiar in the art and could include species referred to as
"minibodies";
and single variable domain "dAbs". Other species still may incorporate means
for
increasing the valency of the modified antibody V-region domain, i.e. species
having
multiple antigen binding sites for example by engineering dimerization domains
(e.g.,
"leucine zippers") or also chemical modification strategies. Moreover, the
term "antibody"
also relates to multimers of scFv such as diabodies, triabodies or
tetrabodies, tandabs,
flexibodies, bispecific antibodies, and chimeric antibodies, all known in the
art. As used
herein, antibodies are considered to also include any bivalent or multivalent
antibodies.
They also include any antibody derivatives and any other derivatives known to
the skilled
person. In some embodiments, the antibody is a polyclonal antibody. In
preferred
embodiments, the antibody is a monoclonal antibody. Further embodiments of
"antibody"
may be taken from WO 2012/072268.
According to the invention, the term "anti-CD4 antibody" refers to an
antibody, which has
the ability to bind to CD4. Preferably herein, the anti-CD4 antibody is an
anti human CD4
antibody. "CD4" or "cluster of differentiation 4" refers to a protein, more
precisely a
surface glycoprotein, well known to the person skilled in the art [cf. above,
cf. also Bowers
et al., 1997]. In the present context CD4 may also refer to a fragment of full-
length CD4,
or an otherwise modified form of CD4, provided that the fragment or otherwise
modified
form still functions as an antigen in the context of the antibody of the
present invention.
Particularly preferred examples of anti-CD4 antibodies that may be used in
accordance
with the present invention are described elsewhere herein.
Non-limiting (further) examples of CD4 antagonists are peptide ligands
(including
naturally occurring peptide ligands and peptide constructs).
Certain further embodiments include aptamers. In a further embodiment, the CD4
antagonist is cyclo(CNSNQIC). In a further embodiment, the CD4 antagonist is
4,4'-
diisothiocyano-2,2'-dihydrostilbenedisulfonic acid.

CA 03012277 2018-07-23
WO 2017/140735 PCT/EP2017/053417
- 15 -
Generally, in preferred embodiments of the invention, the modified graft
comprises a CD4
antagonist.
In further aspects herein, which are related to the first, second and third
aspects, said
method(s) comprise(s) a step of introducing into said subject an unmodified
graft, wherein
said subject has previously been introduced with a modified graft, wherein the
modified
graft is a cell graft containing immune cells, wherein said modification
inhibits CD4. In a
preferred embodiment, said method(s) comprise(s) a step of introducing into
said subject
an unmodified graft, wherein a modified graft has previously been introduced
into said
subject, wherein the modified graft is a cell graft containing immune cells,
wherein said
modification inhibits CD4. Generally, preferred embodiments of those related
aspect
correspond to those of the respective aspects as described herein.
In a fourth aspect herein, the present invention relates to an unmodified
graft, a modified
graft and/or an anti-CD4 antibody for use in any one or more method(s) of the
group
consisting of i) a chirurgical transplantation method, ii) a method of
inducing
transplantation tolerance, wherein a modified graft is used for inducing
transplantation
tolerance to an unmodified graft, iii) a method of transferring one or more
cell
suspension(s), particularly stem cell containing cell suspension(s) from a
donor to a
subject, iv) a method of transferring one or more tissue(s) from a donor to a
subject, v) a
method of transferring one or more partial organ(s) from a donor to a subject,
vi) a method
of transferring one or more organ(s) from a donor to a subject, vii) a method
of transferring
hematopoietic system reconstituting cells from a donor source into a subject,
optionally
followed by transferring one or more organ(s), from said donor source into
said subject,
and viii) a method for enhancing immune reconstitution in a subject,
optionally followed
by transferring one or more organ(s) into said subject.
Generally, the above method(s) preferably comprise(s) a first step of
introducing into said
subject a modified graft, and a second step of introducing into said subject
an unmodified
graft, wherein said modified graft is a cell graft containing immune cells,
wherein said
modification inhibits CD4 (or involves CD4 inhibition, respectively).
Alternatively, in a further variant related to the said fourth aspect, said
method(s)
comprise(s) a step of introducing into said subject an unmodified graft,
wherein said
subject has previously been introduced with a modified graft, wherein the
modified graft is
a cell graft containing immune cells, wherein said modification inhibits CD4
(or involves
CD4 inhibition, respectively). Preferred embodiments of the latter aspect
correspond to
those of the fourth aspect.

CA 03012277 2018-07-23
WO 2017/140735 - 16 - PCT/EP2017/053417
In detail, in one particular embodiment the fourth aspect relates to an
unmodified graft, a
modified graft and/or an anti-CD4 antibody for use in a chirurgical
transplantation method.
In one particular embodiment the fourth aspect relates to an unmodified graft,
a modified
graft and/or an anti-CD4 antibody for use in a method of inducing
transplantation
tolerance, wherein a modified graft is used for inducing transplantation
tolerance to an
unmodified graft.
In one particular embodiment the fourth aspect relates to an unmodified graft,
a modified
graft and/or an anti-CD4 antibody for use in a method of transferring one or
more cell
suspension(s), particularly stem cell containing cell suspension(s) from a
donor to a
subject.
In one particular embodiment the fourth aspect relates to an unmodified graft,
a modified
graft and/or an anti-CD4 antibody for use in a method of transferring one or
more tissue(s)
from a donor to a subject.
In one particular embodiment the fourth aspect relates to an unmodified graft,
a modified
graft and/or an anti-CD4 antibody for use in a method of transferring one or
more partial
organ(s) from a donor to a subject.
In one particular embodiment the fourth aspect relates to an unmodified graft,
a modified
graft and/or an anti-CD4 antibody for use in a method of transferring one or
more organ(s)
from a donor to a subject.
In one particular embodiment the fourth aspect relates to an unmodified graft,
a modified
graft and/or an anti-CD4 antibody for use in a method of transferring
hematopoietic system
reconstituting cells from a donor source into a subject, optionally followed
by transferring
one or more organ(s), from said donor source into said subject.
In one particular embodiment the fourth aspect relates to an unmodified graft,
a modified
graft and/or an anti-CD4 antibody for use in a method for enhancing immune
reconstitution in a subject, optionally followed by transferring one or more
organ(s) into
said subject.
In certain embodiments, the fourth aspect relates to an unmodified graft for
use in any of
said method(s). In certain embodiments, the fourth aspect relates to a
modified graft for use
in any of said method(s). In certain embodiments, the fourth aspect relates to
an anti-CD4
antibody for use in any of said method(s).

CA 03012277 2018-07-23
WO 2017/140735 PCT/EP2017/053417
- 17 -
Generally herein, embodiments of the fourth aspect (and the said variant
related thereto)
correspond to embodiments of the other aspects described herein.
Generally herein, in embodiments of the first, second, third and fourth
aspects herein, the
said first step recited in context with the respective method(s) is carried
out before said
second step recited in context with the said method(s).
In particular embodiments of the first, second, third and fourth aspects
herein, said first
step is carried out from 30 min to 50 years, particularly from one hour to 10
years, such as
to 9, 8, 7, 6, 5, 4, 3, or two years, such as from one day to 1 year, such as
to 11, 10, 9, 8, 7,
6, 5, 4, 3, or two months before said second step. In certain embodiments,
said first step is
carried out from one hour to two months before said second step. In certain
embodiments,
said first step is carried out at least one hour, particularly at least three
weeks, before said
second step.
In particular embodiments, said first step is carried out at least one hour,
particularly at
least one day, particularly at least one month, particularly at least one year
before said
second step. In particular embodiments herein, said first step is carried out
at least 12
hours, particularly at least one day, particularly at least one week,
particularly at least three
weeks, particularly at least six months before said second step.
In particular embodiments, said first step is carried out up to 30 years,
particularly up to 10
years, particularly up to 1 year, particularly up to 1 month before said
second step. In
particular embodiments, said first step is carried out up to 20 years,
particularly up to 5
years, particularly up to 2 years, particularly up to 6 months, particularly
up to 12 weeks
before said second step.
Similarly, in the related further aspects herein, where the said method(s)
herein are defined
as comprising a step of introducing into said subject an unmodified graft,
wherein said
subject has previously been introduced with a modified graft, the timing of
the previous
introduction is preferably further defined as detailed above. Preferably, said
method(s)
herein are defined as comprising a step of introducing into said subject an
unmodified
graft, wherein a modified graft has previously been introduced into said
subject, the timing
of the previous introduction is preferably further defined as detailed above.
Generally, in preferred embodiments of the first, second, third and fourth
aspects herein,
said modified graft comprises stem cells.
In preferred embodiments of the invention, the modified graft is a cell graft
containing
immune cells that has been modified by a CD4 antagonist, particularly wherein
said
modification inhibits CD4.

CA 03012277 2018-07-23
WO 2017/140735 - 18 - PCT/EP2017/053417
Generally, in preferred embodiments of the invention, the modified graft,
particularly a cell
graft containing immune cells, comprises a CD4 antagonist.
In particularly preferred embodiments of the invention, the modified graft is
a cell graft
containing immune cells that comprises an anti-CD4 antibody bound to CD4
epitopes
thereof
In certain preferred embodiments of the invention the modified graft, the
modified graft is
a cell graft containing immune cells that comprises an anti-CD4 antibody bound
to from
40% to 100% of the CD4 epitopes thereof. Such graft is preferably obtainable
by the in
vitro method disclosed herein.
Preferably, the modified cell graft containing immune cells, comprises anti-
CD4 antibodies
bound to from 50% to 100%, particularly 60% to 100%, particularly 70% to 100%,
more
particularly 80% to 100%, more particularly 90% to 100%, more particularly 95%
to
100%, more particularly 99% to 100%, of the accessible CD4 epitopes of said
graft. Most
preferably, essentially all of the accessible CD4 epitopes of the cell graft
containing
immune cells are bound to anti-CD4 antibodies.
In particular embodiments herein, said modified graft is obtained by or
obtainable by an in
vitro method comprising the step a) incubating a cell graft containing immune
cells with an
anti-CD4 antibody, and optionally comprising the step b) removing unbound
antibody
from said graft.
In one preferred embodiment, said modified graft is obtained by or obtainable
by an in
vitro method comprising the step a) incubating a cell graft containing immune
cells with an
anti-CD4 antibody, and optionally comprising the step b) removing unbound
antibody
from said graft, thereby reducing the amount of unbound antibody in said
graft. In a more
preferred embodiment, the amount of unbound antibody in said graft is reduced
by at least
30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or 99%, or is reduced by 100%.
In one preferred embodiment, said modified graft is obtained by or obtainable
by an in
vitro method comprising the step a) incubating a cell graft containing immune
cells with an
anti-CD4 antibody, and optionally comprising the step b) removing unbound
antibody
from said graft by washing the modified graft at least one time or at least 2
times, such as
2, 3, 4, 5 or 6 times. By washing the modified graft, the amount of unbound
antibody in
said graft is reduced. In a more preferred embodiment, the amount of unbound
antibody in
said graft is reduced by at least 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or
99%, or is
reduced by 100%. Suitable washing liquids are known to a skilled person and
include
buffered saline solutions.

CA 03012277 2018-07-23
WO 2017/140735 - 19 - PCT/EP2017/053417
In a preferred embodiment, it is understood that a certain amount of CD4-
positive immune
cells may be lost or removed unspecifically during the washing step(s). In a
yet further
preferred embodiment, the modified graft contains immune cells, in particular
CD4-
positive immune cells, such as CD4 '-T cells, to which anti-CD4 antibody is
bound. In a
more preferred embodiment, the modified graft contains at least 10%, 20%, 30%,
40%,
50%, 60%, 70%, 80%, 90% or 95% of the CD4-positive immune cells of the cell
graft
containing immune cells prior to incubation.
In another preferred embodiment, said modified graft is obtained by or
obtainable by,
respectively, an in vitro method comprising the step a) incubating a cell
graft containing
HI immune cells with an anti-CD4 antibody, and optionally comprising the
step b) removing
only unbound antibody from said graft. Accordingly, the modified cell graft
containing
immune cells may be obtainable in accordance with an in vitro method described
herein.
As used herein, an "in vitro method" refers to a method that is performed
outside a living
subject. It particularly also includes an "ex vivo method", such as in case of
the graft
comprising or being a tissue or an organ, but particularly excludes an "in
vivo method"
performed inside a living subject.
As used herein, "unbound antibody" refers to an antibody which, following the
step of
incubating, is not bound to the graft. In other words, it refers to an
antibody which is not
essentially associated with its ligands on the graft.
Said in vitro method to be used in the invention may be a method of modifying
a cell graft
containing immune cells that comprises the steps of a) incubating a cell graft
containing
immune cells with an anti-CD4 antibody, especially wherein said incubating is
carried out
for from 1 min to 7 days, b) removing unbound antibody from said graft.
Preferably, said
incubating in step a) of said in vitro method is carried out for from 1 min to
1 day.
In one preferred embodiment, said in vitro method to be used in the invention
may be a
method of modifying a cell graft containing immune cells that comprises the
steps of a)
incubating a cell graft containing immune cells with an anti-CD4 antibody,
especially
wherein said incubating is carried out for from 1 min to 7 days, b) removing
unbound
antibody from said graft, thereby reducing the amount of unbound antibody in
said graft. In
a more preferred embodiment, the amount of unbound antibody in said graft is
reduced by
at least 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or 99%, or is reduced by 100%.

Preferably, said incubating in step a) of said in vitro method is carried out
for from 1 min
to 1 day.

CA 03012277 2018-07-23
WO 2017/140735 - 20 - PCT/EP2017/053417
In another preferred embodiment, said in vitro method to be used in the
invention may be a
method of modifying a cell graft containing immune cells that comprises the
steps of a)
incubating a cell graft containing immune cells with an anti-CD4 antibody,
especially
wherein said incubating is carried out for from 1 min to 7 days, b) removing
unbound
antibody from said graft, by washing the modified graft at least one time or
at least 2 times,
such as 2, 3, 4, 5 or 6 times. By washing the modified graft, the amount of
unbound
antibody in said graft is reduced. In a more preferred embodiment, the amount
of unbound
antibody in said graft is reduced by at least 30%, 40%, 50%, 60%, 70%, 80%,
90%, 95%
or 99%, or is reduced by 100%. Preferably, said incubating in step a) of said
in vitro
method is carried out for from 1 min to 1 day.
In a preferred embodiment, it is understood that a certain amount of CD4-
positive immune
cells may be lost or removed unspecifically during the washing step(s). In a
yet further
preferred embodiment, the modified graft obtained by the method contains
immune cells,
in particular CD4-positive immune cells, such as CD4 '-T cells, to which anti-
CD4
antibody is bound. In a more preferred embodiment, the modified graft contains
at least
10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or 95% of the CD4-positive immune
cells of the cell graft containing immune cells prior to incubation.
In another preferred embodiment, said in vitro method to be used in the
invention may be a
method of modifying a cell graft containing immune cells that comprises the
steps of a)
incubating a cell graft containing immune cells with an anti-CD4 antibody,
especially
wherein said incubating is carried out for from 1 min to 7 days, b) removing
only unbound
antibody from said graft. Preferably, said incubating in step a) of said in
vitro method is
carried out for from 1 min to 1 day.
Generally, preferred embodiments for said in vitro methods may be taken from
WO
2012/072268, and are incorporated by reference in their entirety herein.
In step a) of the in vitro method used herein, the (step of) incubating is
preferably carried
out for a time sufficient to allow binding of said antibody to said graft.
Preferably, said
incubating is carried out for a time sufficient to allow the binding of anti-
CD4 antibodies to
from 40% to 100%, particularly 50% to 100%, particularly 60% to 100%,
particularly 70%
to 100%, more particularly 80% to 100%, more particularly 90% to 100%, more
particularly 95% to 100%, more particularly 99% to 100%, of the accessible CD4
epitopes
of said graft. Most preferably, following said incubating, anti-CD4 antibodies
bind to
essentially all of the accessible CD4 epitopes of said graft. In a preferred
embodiment, it is
understood that the accessible CD4 epitopes or CD4-positive immune cells (or
immune
cells bearing the CD4 antigen) of said graft are not removed or substantially
not removed.
Accordingly, in a further preferred embodiment, immune cells bearing the CD4
antigen are

CA 03012277 2018-07-23
WO 2017/140735 - 21 - PCT/EP2017/053417
not depleted, or not substantially depleted from the unmodified graft, and/or
the modified
graft obtained by or obtainable by the in vitro method comprises or
substantially comprises
immune cells bearing the CD4 antigen of the unmodified graft.
An appropriate incubation period will easily be determined by the person
skilled in the art.
Usually, an appropriate incubation period will depend on the type of graft
used. A
preferred incubation period may also dependent on the amount of antibody used.

Generally, where the graft is e.g. a cell suspension, shorter incubation
periods will be
required than where the graft is e.g. an organ. Generally, where the graft
comprises or is a
tissue or an organ, longer incubation periods are preferred to allow the
antibody to be
transported ¨ e.g. via diffusion ¨ into the respective compartments.
Moreover, in any case, the skilled person may easily test the (status of the)
binding of the
anti-CD4 antibodies according to methods well known within the art that may,
for
example, involve flow cytometry.
In certain embodiments, said incubating in step a) of said in vitro method is
carried out for
from 1 min to 7 days. Preferably, said incubating in step a) of said in vitro
method is
carried out for from 1 min to 1 day.
Generally, short incubation periods are preferred herein over long incubation
periods in
order to minimize any possible damage to the graft due to in vitro processing.
Particularly, said incubating may be carried out for from 1 to 150 min,
particularly for
from 5 min to 150 min, more particularly for from 10 min to 150 min, more
particularly for
from 30 min to 150 min, more particularly for from 40 minutes to 120 min, more

particularly for from 45 min to 90 min, especially for from 50 min to 70 min.
In alternative embodiments, said incubating may be carried out for from 150
min to 7 days,
particularly for from 150 min to 5 days, more particularly from 150 min to 3
days, more
particularly from 150 min to 1 day, especially for from 150 min to 8 hours. In
a further
preferred embodiment, incubating may be carried out for from 1 min to 1 day.
As to the "removing" of unbound (anti-CD4) antibody in accordance with e.g.
step b) of
the in vitro method used herein, various ways of performing said step are
known to the
skilled person. One exemplary way of removing unbound antibody from the graft
is by
washing the graft. Washing may e.g. occur by employing centrifugation where
the graft
comprises or is a cell suspension. In one preferred embodiment, the amount of
unbound
antibody in said graft is reduced by washing the graft. In a more preferred
embodiment, the
amount of unbound antibody in said graft is reduced by at least 30%, 40%, 50%,
60%,

CA 03012277 2018-07-23
WO 2017/140735 PCT/EP2017/053417
70%, 80%, 90%, 95% or 99%, or is reduced by 100%. Preferably, only unbound
(anti-
CD4) antibody is removed in accordance with e.g. step b) of the in vitro
method used
herein. A few CD4-positive immune cells may be lost unspecifically during
removal of
unbound antibodies, in particular be washing. In a preferred embodiment, the
modified
.. graft contains at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or 95%
of the
CD4-positive immune cells of the cell graft containing immune cells prior to
incubation.
In said step, preferably at least 40%, more particularly at least 50%, more
particularly at
least 60%, more particularly at least 70%, more particularly at least 80%,
more particularly
at least 90%, of unbound (anti-CD4) antibody are removed from the graft.
Preferably, up to
.. 100% of unbound (anti-CD4) antibody are removed from the graft.
The amount of antibody employed in the above step of incubating is not
particularly
limited. Appropriate amounts may easily be determined by the person skilled in
the art and
may depend on e.g. the type of graft used. Preferably according to the
invention, said
incubating is carried out with an antibody amount of from 0.1 g to 100mg. In a
preferred
amount, Preferably, in accordance with the invention, particularly in case of
the graft being
a cell suspension, an amount of from 2 x 106 cells to 2 x 1010 nucleated
cells, particularly
of from 4 x 106 to 1 x 109 nucleated cells, more particularly of from 1 x 107
to 1 x 108
nucleated cells are administered to said subject, preferably to the human
subject.
accordingly, in a yet further embodiment, said incubating is carried out with
an antibody
amount of from 0.1 g/1 x 109 nucleated cells to 100mg/2 x 106 nucleated cells,
more
preferably 0.1 g/1 x 109 nucleated cells to 100mg/2 x 107 nucleated cells.
In particular embodiments said incubating in step a) of said in vitro method
is carried out
with an antibody amount of from 0.1 g/m1 to 10mg/ml.
In certain embodiments, particularly where the graft is a cell suspension,
said incubating is
carried out with an antibody concentration of from 0.1 g/m1 cell suspension to
150 g/m1
cell suspension, particularly from 7 g/m1 cell suspension to 100 g/m1 cell
suspension,
more particularly from 30 g/m1 cell suspension to 100 g/m1 cell suspension,
especially
from 40 g to 60 g/m1 cell suspension.
.. In some embodiments, particularly where the graft is a tissue or where the
graft is an
organ, said incubating is carried out with an antibody amount of from 0.1mg to
10mg,
particularly from lmg to 10mg, more particularly from 2mg to 9mg, more
particularly
from 3mg to 8mg, especially from 4mg to 6mg.
In some embodiments, particularly where the graft is a tissue or where the
graft is an
organ, said incubating is carried out with an antibody concentration in the
incubation

CA 03012277 2018-07-23
WO 2017/140735 - 23 - PCT/EP2017/053417
solution of from 0.1mg/m1 to 10mg/ml, particularly from 1mg/m1 to 10mg/ml,
more
particularly from 2mg/m1 to 9mg/ml, more particularly from 3mg/m1 to 8mg/ml,
especially
from 4mg/m1 to 6mg/ml. Preferably, the specified volume includes the volume of
said
tissue or organ as well as the volume of the (antibody-containing) solution,
in which said
tissue or organ is incubated.
In some embodiments, particularly where the graft is a tissue or where the
graft is an
organ, said incubating is carried out by incubating said tissue or organ in a
solution having
an antibody concentration of from 10 g/m1 to 150 g/ml, particularly from 20
g/m1 to
100 g/ml, more particularly from 30 g/m1 to 100 g/ml, especially from 40 g/m1
to
60 g/ml. Preferably, the specified volume includes the volume of said tissue
or organ as
well as the volume of the (antibody-containing) solution, in which said tissue
or organ is
incubated.
When incubating tissues and/or organs with a CD4 antagonist (e.g. an anti-CD4
antibody-
containing solution), the skilled person will preferably readily perform such
incubation e.g.
by means of a suitable container.
In any case, the selection of suitable amounts of CD4 antagonist, such as of
anti-CD4
antibody, is well within the expertise of the skilled person. Generally,
higher amounts or
concentrations, respectively, of antagonist (e.g. antibody) are preferred
where the graft
comprises or is a tissue or an organ. Moreover, the selection of an exact
amount or a
.. concentration, respectively, of antagonist (e.g. antibody) used will also
depend on the size
of such tissue or organ.
In the following, further particular embodiments of the modified graft to be
used in the
invention are described:
In some preferred embodiments of the invention, the modified graft comprises
stem cells.
A graft comprising stem cells may also be referred to herein as a stem cell
graft.
According to the present invention, the modified graft may comprise cells
bearing the CD4
antigen. Preferably, the modified graft comprises immune cells, particularly
immune cells
bearing the CD4 antigen. Such cells are well known to the person skilled in
the art. In
certain preferred embodiments, these immune cells are CD4 positive T
lymphocytes or
precursor cells thereof In certain preferred embodiments, these immune cells
include, but
are not limited to T helper cells and cells belonging to the monocyte and
macrophage
lineage, such as monocytes and macrophages. Another example for such cells are

microglia.

CA 03012277 2018-07-23
WO 2017/140735 - 24 - PCT/EP2017/053417
In some embodiments, the modified graft comprises, preferably is, a tissue,
preferably a
stem-cell-containing tissue. According to the present invention, suitable
tissues include,
but are not limited to blood, muscle, adipose tissue, connective tissue,
epithelium,
embryonic, and cellular tissue.
In some embodiments, the modified graft comprises, preferably is, an organ,
preferably a
stem-cell-containing organ. Suitable organs include, but are not limited to
skin, intestine,
kidney, and liver. Preferably, said organ is an intestine.
In preferred embodiments, the modified graft comprises, preferably is, a cell
suspension,
preferably a stem-cell-containing cell suspension. Suitable cell suspensions
and methods
for obtaining them are well known to the skilled person. For example, a cell
suspension
graft may be obtained by puncture of bones comprising bone marrow, e.g.
puncture of the
iliac crests or sterna or taken from stem cell niches throughout the whole
body, e.g. fat
tissue, tooth root, root of a hair and any other source mentioned above.
In preferred embodiments, the modified graft, particularly the cell
suspension, particularly
the stem-cell-containing cell suspension, comprises T cells, monocytes and
macrophages.
In another preferred embodiment, the modified graft, particularly the cell
suspension,
comprises T cells and non-immune cells, such as stem cells. In another
preferred
embodiment, the stem-cell-containing tissue, the stem-cell-containing organ,
or the stem-
cell-containing cell suspension, comprises immune cells, more preferably
immune cells
bearing the CD4 antigen, in particular CD4 positive T lymphocytes or precursor
cells
thereof.
In another preferred embodiment, the graft or cell graft, such as the stem-
cell-containing
tissue, the stem-cell-containing organ, or the stem-cell-containing cell
suspension,
comprises immune cells bearing the CD4 antigen, in particular CD4 positive T
lymphocytes or precursor cells thereof
In certain preferred embodiments the modified graft, particularly the cell
suspension,
comprises any of bone marrow stem cells, peripheral blood stem cells,
umbilical cord
blood stem cells, adult stem cells of the bone marrow such as NA-BMCs,
embryonic stem
cells (particularly derived and/or reprogrammed from adult stem cells), and
any pluripotent
stem cells, wherein the latter includes induced pluripotent cells, such as
cells derived
and/or reprogrammed from adult stem cells (i.e. including pluripotent
embryonic stem
cells).
In preferred embodiments, the modified graft is a bone marrow suspension,
particularly
comprising bone marrow stem cells. Generally, the modified graft, particularly
the bone

CA 03012277 2018-07-23
WO 2017/140735 PCT/EP2017/053417
- 25 -
marrow suspension, may additionally comprise any of stem cells comprised in
blood cells,
cord blood cells, donor lymphocytes, peripheral blood stem cells, adult stem
cells of the
bone marrow, embryonic stem cells (particularly derived and/or reprogrammed
from adult
stem cells), and any pluripotent stem cells, wherein the latter includes
induced pluripotent
cells, such as cells derived and/or reprogrammed from adult stem cells (i.e.
including
pluripotent embryonic stem cells).
The modified graft, particularly the bone marrow suspension, may additionally
comprise
any of stem cells comprised in blood cells, cord blood cells, donor
lymphocytes, peripheral
blood stem cells, and/or adult stem cells of the bone marrow.
Generally, it is intended that the cell suspension also includes any cell
suspension that
comprises (any combination of) stem cells, optionally along with any
(combination of)
other cells.
In preferred embodiments herein, said modified graft is selected from the
group consisting
of a cell suspension containing T cells and monocytes/macrophages the cell
suspension
comprising bone marrow cells, non-adherent bone marrow cells, peripheral blood
cells,
and/or cord blood cells; a cell suspension comprising lymphocytes, monocytes
and/or
macrophages; a stem-cell-containing tissue; a stem-cell-containing organ; an
immune cell
containing tissue; and an immune cell containing organ.
Further embodiments of the unmodified graft herein correspond to the further
particular
embodiments described above for the modified graft.
Generally herein, the amount of cells contained in the graft (e.g. in the
modified graft) is
not particularly limited. Any person skilled in the art will easily be able to
choose
appropriate amounts of a graft and of cells of the graft for transplantation.
Furthermore,
suitable guidance is also available e.g. from the specific guidelines for
transplantation
developed by the "Deutsche Bundesarztekammer", e.g. for human hematopoietic
stem
cells into patients.
In preferred embodiments herein, said modified graft is a cell suspension and
the
respective said method (or use, respectively) comprises administration of an
amount of
from 2x106 cells to 2x1015 nucleated cells to said subject, wherein preferable
amounts will
readily be selected by the skilled person. Exemplary preferred ranges include
from 1x107
to lx1014 nucleated cells, from 1x107 to lx1014 nucleated cells, from 1x108 to
lx 1013
nucleated cells, from 1x109 to lx1012 nucleated cells, from lx101 to lx1011
nucleated
cells, from 1x106 to 1x108 nucleated cells, from 1x107 to 1x109 nucleated
cells, from 1x108
to lx101 nucleated cells, from 1x109 to lx1011 nucleated cells, from lx101
to lx1012

CA 03012277 2018-07-23
WO 2017/140735 - 26 - PCT/EP2017/053417
nucleated cells, from lx1011 to lx1013 nucleated cells, from lx1012 to lx1014
nucleated
cells, and from lx1013 to lx1015 nucleated cells.
Further embodiments of the unmodified graft herein correspond to the said
preferred
embodiments described above for the modified graft.
Where the graft to be used herein comprises or is a tissue or an organ, any
suitable amounts
of said tissue or organ may be administered to said subject. As will be
understood by the
skilled person, cell numbers in tissues or organs are difficult to determine.
Particularly for
this reason, the amount of cells contained in the grafts is not particularly
limited.
Appropriate amounts will easily be determined or selected by the skilled
person, e.g. taking
into consideration the particular type of subject, graft and/or disease to be
treated. In case
of organs, the administration of whole organs is preferred.
In preferred embodiments of first, second, third and fourth aspects herein,
the respective
said method i) implies tolerance or partial tolerance within said unmodified
graft against
the recipient's tissue; and/or ii) implies tolerance or partial tolerance
within the recipient's
tissue against said unmodified graft; and/or iii) implies a reduced likelihood
of developing
any one of the group consisting of GvHD, donor graft rejection, and organ
rejection, upon
translation of the unmodified graft.
Accordingly, in some embodiments herein, the respective said method implies
tolerance or
partial tolerance within said unmodified graft against the recipient's tissue.
Accordingly, in
some embodiments herein, the respective said method implies tolerance or
partial tolerance
within the recipient's tissue against said unmodified graft. Accordingly, in
some
embodiments herein, the respective said method implies a reduced likelihood of

developing any one of the group consisting of GvHD, donor graft rejection, and
organ
rejection, upon transplantation of the unmodified graft.
In some embodiments herein, the use of the modified graft implies a reduced
likelihood of
developing any one of the group consisting of GvHD, donor graft rejection, and
organ
rejection; particularly of GvHD, upon transplantation of said graft. In other
embodiments,
the use implies tolerance within the transplanted immunocompetent cells
against the
recipient's tissue upon transplantation of said modified graft. In other
embodiments, the
use implies tolerance against the modified graft upon transplantation of said
modified
graft. In other embodiments, the use implies tolerance or partial tolerance
within the
recipient's tissue against the modified graft upon transplantation of said
modified graft. In
other embodiments, the use is for silencing cell activation within said graft.
In a preferred
embodiment, the use is for reducing the CD4-positive immune cells' ability to
be activated
within said graft. In a preferred embodiment, the use is for obtaining anergy
of the CD4-

CA 03012277 2018-07-23
WO 2017/140735 PCT/EP2017/053417
- 27 -
positive immune cells within said graft. In preferred embodiments, the use
implicates / is
for any combination of the above.
In some preferred embodiments herein, the use of the modified graft results in
a reduced
likelihood of developing any one of the group consisting of GvHD, donor graft
rejection,
and organ rejection; particularly of GvHD, upon transplantation of said graft.
In other
preferred embodiments, the use results in tolerance of the transplanted
immunocompetent
cells against the recipient's tissue upon transplantation of said modified
graft. In other
preferred embodiments, the use results in tolerance against the modified graft
upon
transplantation of said modified graft. In other preferred embodiments, the
use results in
tolerance or partial tolerance of the recipient's tissue against the modified
graft upon
transplantation of said modified graft. In other preferred embodiments, the
use is for
preventing cell activation or reducing the cells' ability to be activated
within said graft. In a
preferred embodiment, the use is for reducing the CD4-positive immune cells'
ability to be
activated within said graft. In a preferred embodiment, the use is for
obtaining anergy of
the CD4-positive immune cells within said graft. In preferred embodiments, the
use
implicates / is for any combination of the above.
In the present invention, generally, the said subject (or recipient,
respectively) may be
allogeneic or xenogeneic with respect to the donor(s).
As will readily be understood by the skilled person, the recipient and the
respective
donor(s) are referred to as being allogeneic when they are separate
individuals of the same
species, whereas the recipient and the respective donor(s) are referred to as
being
xenogeneic when they are derived from different species.
Accordingly, in certain preferred embodiments herein, the said subject is
allogeneic with
respect to the donor of the said unmodified graft and to the donor of the said
modified
graft. In other embodiments herein, the said subject is xenogeneic with
respect to the donor
of the said unmodified graft and to the donor of the said modified graft.
Similarly, in the present invention, generally, the donors may be allogeneic
or xenogeneic
with respect to each other.
As will readily be understood by the skilled person, the donor(s) are referred
to as being
allogeneic when they are separate individuals of the same species, whereas the
donor(s) are
referred to as being xenogeneic when they are derived from different species.
Accordingly, in certain embodiments herein, the donor of the modified graft is
xenogeneic
with respect to the donor of the said unmodified graft. In certain preferred
embodiments

CA 03012277 2018-07-23
WO 2017/140735 - 28 - PCT/EP2017/053417
herein, the donor of the modified graft is allogeneic with respect to the
donor of the
unmodified graft.
In the present invention, the nature of and relationship between the donors of
the modified
graft and the unmodified graft and the recipient are not particularly limited.
Hence, in
certain non-limiting examples, and e.g. where the donor(s) and recipient are
mice, the
donor(s) may comprise full HLA mismatches with respect to the recipient.
Hence, in
certain preferred non-limiting examples, and e.g. where the donor(s) and
recipient are
mice, the donor(s) may have full HLA mismatches with respect to the recipient.
In other non-limiting examples, the donor(s) may e.g. be haploidentical (such
as
transplantation between parents and infants) to the recipient and may e.g. be
HLA-partially
matched family members, parents, siblings or children of the recipient.
In any case, it is preferred in the present invention that the donor(s) and
the recipient (or
subject, respectively) are HLA-related.
Likewise, in the present invention, the nature of and relationship between the
donors of the
modified graft and the unmodified graft are not particularly limited. Hence,
in certain non-
limiting examples, the donor(s) may comprise, preferably have, full HLA
mismatches with
respect to each other. In other non-limiting examples, one donor may e.g. be
haploidentical
to the other donor, and may e.g. be HLA-partially matched family members,
parents,
siblings or children of the other donor.
In any case, it is preferred in the present invention that the modified graft
and the
unmodified graft are from HLA-related donors. Accordingly, the respective
donors in
context with the present invention are preferably HLA-related (i.e. HLA-
related to each
other).
As used herein, the term "HLA-related donors" is preferably understood as
referring to
donors comprising, preferably having, not more than 50% HLA mismatches. In
preferred
embodiments, the donors comprise, preferably have, not more than 40% HLA
mismatches,
preferably not more than 30% HLA mismatches, preferably not more than 20% HLA
mismatches, preferably not more than 10% HLA mismatches, preferably no HLA
mismatch.
Similarly, a given donor and recipient are referred to as being "HLA-related"
when they
comprise, preferably have, not more than 50% HLA mismatches. In preferred
embodiments, a given donor and recipient comprise, preferably have, not more
than 40%
HLA mismatches, preferably not more than 30% HLA mismatches, preferably not
more

CA 03012277 2018-07-23
WO 2017/140735 - 29 - PCT/EP2017/053417
than 20% HLA mismatches, preferably not more than 10% HLA mismatches,
preferably
no HLA mismatch.
Accordingly, and particularly in case of humans, the term "HLA-related" is
preferably
understood as referring to the presence of not more than 50% HLA mismatches,
preferably
not more than 40% HLA mismatches, preferably not more than 30% HLA mismatches,
preferably not more than 20% HLA mismatches, preferably not more than 10% HLA
mismatches, preferably no HLA mismatch.
Accordingly, and particularly in case of humans, the term "HLA-related" is
preferably
understood as referring to the presence of not more than six HLA mismatches,
preferably
not more than five HLA mismatches, preferably not more than four HLA
mismatches,
preferably not more than three HLA mismatches, preferably not more than two
HLA
mismatches, preferably not more than one HLA mismatch, preferably to no HLA
mismatch.
In certain preferred embodiments, in case of humans, the said percentages and
or numbers
of mismatch(es) are determined with respect to the HLA loci HLA-A, -B, C-, -
DR, -DQ,
and -DP (corresponding to a total of 12 HLA loci). In other preferred
embodiments, the
said mismatch(es) are determined with respect to the HLA loci HLA-A, -B, C-, -
DR, and -
DQ (corresponding to a total of 10 HLA loci). In other preferred embodiments,
the said
mismatch(es) are determined with respect to the HLA loci HLA-A, -B, C-, and -
DR
(corresponding to a total of 8 HLA loci).
Generally herein, methods for determining HLA mismatches are not particularly
limited,
but are readily available and apparent to the person skilled in the art.
Furthermore, as will be readily understood by the skilled person, in the above
definitions of
the term "HLA related", HLA loci may be replaced by corresponding appropriate
loci
where non-human donors and recipients are compared to each other. Said loci
are readily
known to the skilled person.
Moreover, further envisaged herein are cases where the modified graft (e.g.
without or
after genetic manipulation) is HLA-related to the unmodified graft and vice
versa. Hence,
in certain preferred embodiments of the present invention, the modified graft
and the
unmodified graft are "HLA-related", wherein particular embodiments for "HLA-
related"
correspond to any of those above. Moreover, further envisaged herein are cases
where the
modified graft and/or the unmodified graft (e.g. without or after genetic
manipulation) is
HLA-related to the recipient. Hence, in certain preferred embodiments of the
present
invention, the modified graft is HLA-related to the recipient. Hence, in
certain preferred

CA 03012277 2018-07-23
WO 2017/140735 - 30 - PCT/EP2017/053417
embodiments of the present invention, the unmodified graft is HLA-related to
the recipient.
Again, particular embodiments for "HLA-related" correspond to any of those
above.
Accordingly, as will also be understood by the skilled person, the modified
and unmodified
grafts may be HLA-related in case the respective donors are not HLA-related ¨
and a given
modified graft and/or unmodified may be HLA-related to a recipient in case the
respective
donor(s) and recipient are not HLA-related. Furthermore, as will also be
understood by the
skilled person, (grafts or) donors may also be HLA-related in case the donors
are
xenogeneic ¨ and (modified and unmodified grafts or) a given donor and
recipient may
also be HLA-related in case the donor and recipient are xenogeneic.
To the latter end, e.g. (one of) the donor(s) (or grafts, respectively) may be
genetically
manipulated to achieve one or more HLA matches or further HLA matches.
Accordingly,
in certain preferred embodiments, donor(s) with respect to the recipient (or
the donors of
the modified graft and the unmodified graft, respectively) preferably
comprise, preferably
have, not more than six HLA mismatches after genetic manipulation e.g. of
donor cells,
preferably comprise, more preferably have, not more than three HLA mismatches
after
genetic manipulation e.g. of donor cells, preferably comprise, more preferably
have, not
more than two HLA mismatches after genetic manipulation e.g. of donor cells,
preferably
comprise, more preferably have, not more than one HLA mismatch after genetic
manipulation e.g. of donor cells, preferably comprise, more preferably have,
no HLA
mismatch after genetic manipulation e.g. of donor cells. Further embodiments
in the latter
context correspond to those as described above.
As also described above, in the present invention, the modified graft and the
unmodified
graft are preferably from HLA-related donors or, in other words, the donor of
the modified
graft and the donor of the unmodified graft are preferably HLA-related.
Non-limiting particular examples of HLA-related donors include tissue-type
matched
donors and related twins. Hence, in certain embodiments, said HLA related
donors are
tissue-type matched donors (such as a tissue HLA type matched donor).
Accordingly, in
certain embodiments, the modified graft and the unmodified graft are from
tissue-type
matched donors. In certain embodiments, said HLA related donors are related
twins,
especially monozygotic twins. Accordingly, in certain embodiments, the
modified graft
and the unmodified graft are from related twins, especially from monozygotic
twins.
Generally, in particularly preferred embodiments, the modified and unmodified
grafts are
from the same donor. Accordingly, in preferred embodiments, the said HLA-
related donors
may be one and the same donor. Hence, the term HLA-related donors as used
herein may
actually refer to one (particular) donor. Hence, in particular preferred
embodiments, said

CA 03012277 2018-07-23
WO 2017/140735 - 31 - PCT/EP2017/053417
HLA related donors are one particular donor. Hence, in certain preferred
embodiments of
the present invention, both the modified graft and the unmodified graft are
from the same
donor.
In preferred embodiments of the invention, the anti-CD4 antibody i) is
selected from the
group consisting of Max16H5, OKT4A, OKTcdr4a, cMT-412, YHB.46, particularly
wherein said anti-CD4 antibody is Max16H5; or ii) is antibody 30F16H5; or iii)
is
obtainable from a cell line deposited with accession number ECACC 88050502; or
iv) is
obtainable from a cell line MAX.16H5/30F16H5 deposited with the DSMZ on
December
2, 2011; or v) is antibody 16H5.chimIgG4; or vi) is obtainable from a cell
line
CD4.16H5.chimIgG4 deposited with the DSMZ on December 2, 2011; or vii) is an
antibody comprising the VH and the VK of antibody 16H5.chimIgG4; or viii) is
an
antibody comprising a VH and a VK of an antibody obtainable from a cell line
CD4.16H5.chimIgG4 deposited with the DSMZ on December 2, 2011; or ix) is an
antibody comprising any combination of a VH selected from SEQ ID NOs: 1-10 and
of a
VK selected from SEQ ID NOs: 11-20, particularly wherein said combination is
selected
from VH1NK1, VH2NK2, VH4NK2 and VH4NK4, especially wherein said
combination is VH2NK2, or x) is a mixture of antibodies selected from the
antibodies
according to i) to ix) above.
Accordingly, referred anti-CD4 antibodies for use in accordance with the
present invention
are selected from the group consisting of Max16H5, OKT4A, OKTcdr4a, cMT-412,
YHB.46. A particularly preferred anti-CD4 antibody is Max 16H5. Cells for the
production
of Maxl6H5 have been deposited with the ECACC (European Collection of Cell
Cultures)
with accession number ECACC 88050502. Said antibody is also disclosed in DE
3919294,
which is incorporated by reference herein. As used herein, the antibody
"Max16H5" may
also be referred to as "Max.16H5", "MAX16H5" or "MAX.16H5", or also "30F16H5"
(wherein the latter name is also the name of deposited cells producing said
antibody).
Max.16H5 may also be obtained from the cell line MAX.16H5/30F16H5 (cf. deposit
DSM
ACC3148). Another particularly preferred anti-CD4 antibody for use in the
invention is
16H5.chimIgG4. As used herein, said antibody may also be referred to as
"16H5.chim" or
as "CD4.16H5.chimIgG4" (wherein the latter name is also the name of deposited
cells
producing said antibody). 16H5.chimIgG4 may be obtained from the cell line
CD4.16H5.chimIgG4 (cf. deposit DSM ACC3147).
In detail, certain preferred anti-CD4 for use in the present invention are
e.g. obtainable
from any of the following deposits of biological material: i) deposit with the
European
Collection of Cell Cultures having the accession number ECACC 88050502 (which
is e.g.
also described in application DE 3919294); ii) deposit "MAX.16H5/30F16H5",
accession

CA 03012277 2018-07-23
WO 2017/140735 - 32 - PCT/EP2017/053417
number DSM ACC3148, deposited with the DSMZ on December 2, 2011; iii) deposit
"CD4.16H5.chimIgG4", accession number DSM ACC3147, deposited with the DSMZ on
December 2, 2011. All of these deposits involve cells or cell lines,
respectively, from
which particular anti-CD4 antibodies in the context with the present invention
may be
obtained.
The present invention further relates to alternative embodiments of
embodiments disclosed
herein, where the term "ECACC 88050502" is replaced by "MAX.16H5/30F16H5".
Likewise, the present invention further relates to embodiments, where the term
"cell line
ECACC 88050502" as used herein, or an equivalent term, is replaced by the term
"cell line
MAX.16H5/30F16H5" or an equivalent term. The present invention further relates
to
alternative embodiments of embodiments disclosed herein, where the term "ECACC

88050502" is replaced by "CD4.16H5.chimIgG4". Likewise, the present invention
further
relates to embodiments, where the term "cell line ECACC 88050502" as used
herein, or an
equivalent term, is replaced by the term "cell line CD4.16H5.chimIgG4" or an
equivalent
term.
In further embodiments of the present invention, particular anti-CD4
antibodies (including
modified antibodies, etc.) that may be used correspond to the anti-CD4
antibodies
described in patent application W02012/072268 (cf. particularly the
"additional aspect"
described therein), which document is incorporated by reference in its
entirety herein. This
particularly also applies to the embodiments according to items 1-33 disclosed
on pages 42
et seqq. of W02012/072268.
Likewise, particular (embodiments for) methods to obtain / prepare further
anti-CD4
antibodies that may be used in embodiments of the present invention correspond
to those
described in W02012/072268 as well as the references disclosed therein.
Likewise,
particular (embodiments for) methods to evaluate anti-CD4 antibodies
(including e.g.
method for the measurement of the affinity of antibodies) that may be used in
embodiments of the present invention correspond to those described in
W02012/072268 as
well as the references disclosed therein.
Herein, the term "VH" generally refers to the heavy chain variable region of
the heavy
chain of an antibody. The "heavy chain variable region" is also referred to as
"heavy chain
immunoglobulin variable domain". Also these terms are well-known in the art.
The term
"VL" generally refers to the light chain variable region of the light chain of
an antibody.
The "light chain variable region" is also referred to as "light chain
immunoglobulin
variable domain". These terms are well-known in the art, too. VH preferably
means a

CA 03012277 2018-07-23
WO 2017/140735 - 33 - PCT/EP2017/053417
polypeptide that is about 110 to 125 amino acid residues in length. Similarly,
VL
preferably means a polypeptide that is about 95-130 amino acid residues in
length.
In preferred embodiments herein, prior to introduction into said subject, said
unmodified
graft and/or modified graft is/are additionally incubated with soluble
bioactive molecules.
Accordingly, in preferred embodiments herein, prior to introduction into said
subject, said
unmodified graft is additionally incubated with soluble bioactive molecules.
Accordingly,
in preferred embodiments herein, prior to introduction into said subject, said
modified graft
is additionally incubated with soluble bioactive molecules.
In each case, in particular such bioactive molecules are agents promoting
immunosuppression, immunotolerance and/or formation of regulatory T cells.
Preferably,
each case, such bioactive molecules are agents promoting immunosuppression,
immunotolerance and/or (favored) formation and/or (favored) activation of
regulatory T
cells.
Preferred exemplary agents are selected from the group consisting of IL-2, TGF-
13,
rapamycin, retinoic acid, 4-1BB ligand, and anti-CD28 antibodies; or any
combination
thereof.
Generally, the grafts for use in the present invention may optionally be
administered to the
subject together with any medicament or combination of medicaments. This
applies to both
the modified and unmodified grafts.
Said medicament(s) may be administered prior to, together with and/or
following
transplantation. Suitable administration modes and routes are not particularly
limited and
will easily be chosen by the skilled person. Preferably, such medicament(s)
support the
features or advantages, respectively, of the present methods, uses, modified
grafts, or
modified grafts for use described hereinabove, such as reducing the likelihood
of any one
of the group consisting of GvHD, donor graft rejection, and organ rejection.
Non-limiting
examples for such medicaments include rapamycin and retinoic acid.
In preferred embodiments herein, the modified graft is used i) for achieving
tolerance or
partial tolerance of the modified graft against the recipient's tissue; and/or
ii) for achieving
tolerance or partial tolerance of the recipient's tissue against the modified
graft; and/or iii)
for reducing the likelihood of any one of the group consisting of GvHD, donor
graft
rejection, and organ rejection upon transplantation of said modified graft.
In preferred embodiments herein, the modified graft is used i) for achieving
tolerance or
partial tolerance of the unmodified graft against the recipient's tissue;
and/or ii) for

CA 03012277 2018-07-23
WO 2017/140735 - 34 - PCT/EP2017/053417
achieving tolerance or partial tolerance of the recipient's tissue against the
unmodified
graft; and/or iii) for reducing the likelihood of any one of the group
consisting of GvHD,
donor graft rejection, and organ rejection upon transplantation of said
unmodified graft.
In a further aspect, the invention features methods of treating a subject in
need of such
treatment in accordance with the used described herein. In preferred
embodiments, said
grafts, subjects, methods, and/or diseases are as described hereinabove.
In a further aspect, the invention features an unmodified graft, a modified
graft and/or an
anti-CD4 antibody for use in the manufacture of a medicament for the treatment
of one or
more diseases treatable by transplantation in a subject and/or for any of the
(medical) uses
described herein. In preferred embodiments, said use is further defined as
described herein
above. Accordingly, in preferred embodiments, said grafts, subjects, methods,
and/or
diseases are as described hereinabove.
In an even further aspect, the graft (particularly the modified graft) for use
in the present
invention, may or may not comprise stem cells. That is, according to the
latter aspect, the
cell graft containing immune cells may be replaced by any graft and includes a
graft
comprising stem cells as well as a graft not comprising stem cells. In other
words, the graft
of the invention or used in accordance with the invention may be any graft or
may be a cell
graft containing immune cells. In even other words, in certain embodiments a
graft used in
accordance with the invention comprises stem cells, whereas in other
embodiments, a graft
used in accordance with the invention does not comprise stem cells. In certain

embodiments, the graft does not comprise isolated CD4 ' cells. In certain
embodiments, the
graft does not comprise purified D0.11.10 CD4 ' T cells.
Generally, further embodiments of the various aspects herein correspond to
those described
in W02012/072268.
Generally, as to further mechanistic, experimental and theoretical
characteristics related to
(side) aspects of the present invention, reference is also made to
W02012/072268.
Generally, the invention also relates to embodiments, where the term
"comprises" or an
equivalent term is replaced by "has" or an equivalent term. For example, the
invention
generally also relates to embodiments, where the term "comprising" or an
equivalent term
.. is replaced by "having" or an equivalent term.
In the following, the present invention is further illustrated by examples
which are not
intended to limit the scope of the present invention.
EXAMPLES

CA 03012277 2018-07-23
WO 2017/140735 PCT/EP2017/053417
- 35 -
Example 1:
Animals and experimental design
Donor triple transgenic mice (human CD4'/', murine CD4', HLA-DR3) TTG-057B1/6
were bred at the Animal Facility at the University Leipzig [Fricke, 2014;
Schmidt, 2015].
Recipient Balb/cwt mice were purchased from Charles River (Sulzfeld, Germany;
http://jaxmice.jax.org) [Fricke, 2014; Schmidt, 2015]. All mice were
maintained under
standardized conditions [Fricke, 2014; Schmidt, 2015]. In transgenic mice, the
CD4
transgene includes its own promoter ligated to a murine CD4 enhancer element
thus
leading to T cell subset-specific expression [Fricke, 2014; Schmidt, 2015].
Murine CD8 '
cells are not affected [Fricke, 2014; Schmidt, 2015]. Transgenic mice express
the HLA-
DR3 molecule in addition to the murine MHC II complex [Fricke, 2014; Schmidt,
2015].
The TTG-057B1/6 mice have complete functional murine immune systems [Fricke,
2014;
Schmidt, 2015]. The mice were fed ad libitum [Fricke, 2014; Schmidt, 2015].
All mice
were housed, treated, or handled in accordance with the guidelines of the
University
Leipzig Animal Care Committee and were approved by the Regional Board of
Animal
Care for Leipzig [Fricke, 2014; Schmidt, 2015].
Allogeneic hematopoietic stem cell grafts from donor TTG-057B1/6 were
incubated with
MAX.16H5 IgGi or control antibodies for 2 hours before transplantation into
Balb/cwt
[Fricke, 2014; Schmidt, 2015]. Survival and GvHD occurrence were measured
daily,
engraftment, hematological (WBC subsets) and immunological reconstitution
(murine
CD4, human CD4, murine CD8, HLA-DR3, and H2Kb) were measured weekly [Fricke,
2014; Schmidt, 2015].
Irradiation protocol
For the irradiation of mice, the X-Ray apparatus (D3225, Orthovoltage, Gulmay
Medical,
Camberley, UK) was adjusted for animal irradiation as we described previously
[Fricke,
2009]. Irradiation was performed before transplantation of recipient mice.
Preparation of bone marrow cells, splenocytes, and antibody incubation
As isotype control, the antibody from LEAFTM Purified Mouse IgGi (K, Isotype
Ctrl
Antibody, San Diego, CA 92121) was used [Fricke, 2014]. MAX.16H5 IgGi
antibodies
were used as described previously [Fricke, 2014]. Bone marrow cells (BM) and
splenocytes were prepared as described elsewhere [Fricke, 2009; Fricke, 2010;
Fricke,
2011; Fricke, 2012, Fricke, 2014, Schmidt, 2015]. For antibody incubation, the
calculated
amount of antibodies was diluted before use to a final concentration of 1mg/m1
in DMEM

CA 03012277 2018-07-23
WO 2017/140735 - 36 - PCT/EP2017/053417
without FCS [Fricke, 2014]. Subsequently, 1.4x108 of BM and 1.4x108 of
splenocytes
from donors were incubated with 800 g MAX.16H5 IgGi in 15m1 DMEM without FCS
at
room temperature in the dark for 2 hours [Fricke, 2014]. As control, BM and
splenocytes
from the donors without antibody pre-incubation were prepared under the same
conditions
[Fricke, 2014]. After 2 hours of incubation, cells were centrifuged at 300g
for 10 min to
pellet them and washed once in PBS (1x) at 300g for 10 min to remove unbound
antibodies
[Fricke, 2014].
Cell Transplantation
For co-transplantation experiments, 2x107 pre-incubated bone marrow cells of
donor TTG-
C57B1/6 mice were added to 2x107 MAX.16H5 IgGi pre-incubated splenocytes
[Fricke,
2014]. The cell concentration was adjusted to a final volume of 150 1 sterile
0.9% NaCl
[Fricke, 2014]. Subsequently, the grafts were allogeneically transplanted by
intravenous
injection into the lateral tail vein of lethally irradiated recipient Balb/cwt
mice [Fricke,
2014]. Survival, GvHD symptoms, and weight were assessed every day after
transplantation [Fricke, 2014].
Flow cytometry
Before and after transplantation, donor TTG-057B1/6 and recipient Balb/cwt
mice were
analyzed by flow cytometry. For cytometric analysis, cells were prepared and
incubated as
previously described [Fricke, 2009; Fricke, 2010; Fricke, 2011; Fricke, 2012,
Fricke,
2014]. Additionally, the viability of splenocytes and bone marrow cells was
tested before
transplantation by staining with 7-Amino-Actinomycin D (7-AAD). 1x106 cells
were
incubated with 5 1 (0.25 g/test) of 7-AAD in 300 1 PBS at room temperature for
30 min
and measured immediately. Before and after transplantation, recipient Balb/cwt
mice were
analyzed by flow cytometry and the full blood cell counts were determined. The
following
antibodies were used: murine CD4-PECy7, MHC-I (H-2D[b]-PE, and murine CD8-
PerCP
[BD Biosciences, Heidelberg, Germany]; murine CD3-FITC and human CD4-APC
[Beckman Coulter, Krefeld, Germany]; and human HLA-DR3-FITC [Immunotools,
Friesoythe, Germany]. For cytometric analysis of murine FoxP3, the murine Treg
Detection
Kit (Miltenyi Biotec GmbH, Bergisch Gladbach, Germany) was used according to
the
manufacturer's protocol. The data were acquired on a BD FACSCantoTM II Flow
Cytometer and analyzed using BD FACSDivaTM software (both BD Biosciences,
Heidelberg, Germany) [Fricke, 2014].
Skin transplantation

CA 03012277 2018-07-23
WO 2017/140735 PCT/EP2017/053417
- 37 -
For allogeneic skin transplantation Balb/c' t (TTG) mice (showing C57B1/6
blood
chimerism) and Balb/c' t mice were used as recipient and CD4/CD3 mice (TTG) as
donors.
Recipient mice were anesthetized by a standard protocol approved by the
authorities. Skin
of the donor mice from the tails was immediately prepared. The fur on the
shoulder of
recipient mice was shaved carefully before transplantation. Full-thickness
grafts of 5x5mm
donor tail skin (TTG) were transplanted on Balb/c (TTG) or Balb/c' t
recipients.
Transplanted skin graft was secured by surgical sterile thread and
additionally dressed with
adhesive sterile bandage. Recipient mice were awakened by a standard protocol
approved
by the authorities. After surgery, mice were individually housed to avoid
additional
injuries. All procedures of skin transplantation took place under sterile
conditions in an
operating room and were approved by the authorities. Mice were monitored
daily. Fifty
days (50 days) after skin transplantation, the tissue was collected for
histological analysis.
Histology of recipient mice
Histology was done as described previously [Fricke, 2011].
Statistical Analysis
All data are presented as means standard deviation [Fricke, 2012].
Statistical analysis
and graphic presentations were made using SigmaPlot 10.0/SigmaStat 3.5
software
(SYSTAT, Erkrath, Germany) [Fricke, 2012] and GraphPad Prism 5 (v5.03,
GraphPad
Software Inc, Dan Diego, California). P-values calculated with SigmaPlot
11.0/SigmaStat
3.5 software. Test variables were the GvHD score, weight, survival, and cell
numbers.
Analysis of survival curves was done using the log-rank test, analysis of
other parameters
with t-tests, the Mann¨Whitney U test or the Holm-Sidak test [Fricke, 2009].
Results: (cf. also Fig. 1)
It was hypothesized that solid organs (e.g. skin) from third party TTG donor
mice on
Balb/c (TTG) recipient mice, which showed immune tolerance, would not be
rejected after
transplantation. For this, 5 Balb/c (TTG) mice (showing C57B1/6 blood
chimerism) and 4
Balb/c' t (control) were transplanted using tail skin of TTG mice. It had to
be mentioned
that Balb/c (TTG) recipients were about 6 months older as control mice because
of
hematopoietic stem cell transplantation before. Sixty percent (60%) of skin
transplanted
Balb/c (TTG) mice survived and 100% of Balb/c' t mice. Nevertheless, all
surviving Balb/c
(TTG) mice (showing C57B1/6 blood chimerism, N=3), which received a third
party skin
graft from TTG mice did not show graft rejection unlike the controls (Fig. 1).
Graft failure
of TTG skin was observed in the Balb/c' t control group (N=4).
Example 2:

CA 03012277 2018-07-23
WO 2017/140735 - 38 - PCT/EP2017/053417
As to further antibodies for use in the present invention, chimeric anti-CD4
antibodies may
be made, and modified anti-CD4 antibodies may be designed and generated in
accordance
with Example 2 of W02012/072268, which is also incorporated in its entirety by
reference
herein.
Example 3:
Furthermore, as to an animal model for the human immune system that may
supplementarily be used by the skilled person in context with certain issues
of this
invention, reference is made to WO 2006/122545 including all family members of
the
respective patent family such as EP 1887859 and US 2008216182, all of which
are
.. explicitly incorporated by reference herein.

CA 03012277 2018-07-23
WO 2017/140735 - 39 - PCT/EP2017/053417
LIST OF REFERENCES (all of which are explicitly incorporated by reference
herein)
DE 3919294
WO 2006/122545
WO 2012/072268
Barnard, C.N. The operation. A human cardiac transplant: an interim report of
a successful
operation performed at Groote Schuur Hospital, Cape Town. S Afr Med J. 41,
1271-1274
(1967).
Becker, C., Bopp, T. & Jonuleit,H. Boosting regulatory T cell function by CD4
stimulation
enters the clinic. Front Immunol. 3. 164 (2012).
Bellone, M. Autoimmune Disease: Pathogenesis. Encyclopedia of Life Sciences
John Wiley
& Sons, 1-8 (2005).
Bowers, K., Pitcher, C. & Marsh, J.M. CD4:A co-receptor in the immune response
and HIV
infection.. Int. J. Biochem. Cell Biol.. 29, 871-875 (1997).
Calne, R.Y. et al. A study of the effects of drugs in prolonging survival of
homologous
renal transplants in dogs. Ann N Acad Sci. 99, 743-761 (1962).
Carrel, A. & Guthrie, C.C. Successful Transplantation of Both Kidneys from a
Dog into a
Bitch with Removal of Both Normal Kidney from the Latter. Science 23, 394-395
(1906).
Chatenoud, L., Waldmann, H., & Emmrich,F. Tolerance induction in the adult:
'danger at
Le Bischenberg. Immunol. Today 16, 121-123 (1995).
Coligan et al., Current Protocols in Immunology, John Wiley & Sons 1991-1997
Cooke KR, Kobzik L, Martin TR, Brewer J, Delmonte J, Crawford JM et al. An
experimental
model of idiopathic pneumonia syndrome after bone marrow transplantation .1.
The roles of
minor H antigens and endotoxin. Blood 88(8):3230-3239 (1996)
Cooke, K.R. et al. Tumor necrosis factor- alpha production to
lipopolysaccharide
stimulation by donor cells predicts the severity of experimental acute graft-
versus-host
disease. J. C/in. Invest 102, 1882-1891 (1998).
Cooley, D.A. et al. Organ transplantation for advanced cardiopulmonary
disease. Ann
Thorac Surg. 8, 30-46 (1969).
Dausset, J. Iso-leuco-anticorps. Acta Haematol. 20, 1956-1966 (1958).
De Castello, A. & Sturli, A. Ueber die Isoagglutinine im Serum gesunder und
kranker
Menschen. Mfinch med Wschr. 49, 1090-1095 (1902).
De Groot, A.S. & Scott, D.W. Immunogenicity of protein therapeutics. Trend
Immunol. 28,
482-490 (2007).
Durand, J.K. & Willis, M.S. Karl Landsteiner, MD: Transfusion Medicine.
Laboratory
Medicine 41, 53-55 (2009).
Eigler, F.W. Zur Geschichte der Nierentransplantation in Deutschland.
Zentralbl Chir. 127,
1001-1008 (2002).
Emmrich, F. et al. An anti-CD4 antibody for treatment of chronic inflammatory
arthritis.
Agents Actions Suppl 32, 165-170 (1991a).
Emmrich, J., Seyfarth, M., Fleig, W.E., & Emmrich, F. Treatment of
inflammatory bowel
disease with anti-CD4 monoclonal antibody. Lancet 338, 570-571 (1991b).

CA 03012277 2018-07-23
WO 2017/140735 - 40 - PCT/EP2017/053417
Eurotransplant. Total numbers of organ transplantations in Germany in the
period from
1963 to 2013 according to organs. 2015a [cited on June 19, 2015, URL:
http ://de.statista.com/statistik/daten/studie/202037/umfrage/anzahl-der-
organtransplantationen-in-deutschland/ ]
Eurotransplant. 2015b [cited on June 19, 2015, URL:
https://www.eurotransplant.
org/cms/index.php?page=pat germany_
Fricke, S. et at. Allogeneic non-adherent bone marrow cells facilitate
hematopoietic
recovery but do not lead to allogeneic engraftment. PLoS. One. 4. e6157
(2009).
Fricke, S. et at. Characterization of murine non-adherent bone marrow cells
leading to
recovery of endogenous hematopoiesis. Cell Mol. Life Sd. 67, 4095-4106 (2010).
Fricke, S. Measurement and illustration of immune interaction after stem cell
transplantation. Methods Mol. Biol. 690. 315-332 (2011).
Fricke, S. et at. Allogeneic bone marrow grafts with high levels of CD4(+)
CD25(+)
FoxP3(+) T cells can lead to engraftment failure. Cytometry A 81, 476-488
(2012).
Fricke, S. et at. Prevention of graft-versus-host-disease with preserved graft-
versus-
leukemia-effect by ex vivo and in vivo modulation of CD4 ' T-cells. Cell Mol
Life Sci. 71,
2135-2148 (2014).
Harding, S., Lipp, P., & Alexander, D.R. A therapeutic CD4 monoclonal antibody
inhibits
TCR-zeta chain phosphorylation, zeta-associated protein of 70-kDa Tyr31 9
phosphorylation, and TCR internalization in primary human T cells. J. Immunol.
169, 230-
238 (2002).
Harrison, S.C. CD4: Structure and Interactions of an Immunoglobulin
Superfamily
Adhesion Molecule. Acc Chem. Res. 26, 449-453 (1993).
Hsu, D.C. & Katelaris, C.H. Long-term management of patients taking
immunosuppressive
drugs. Australian Prescriber 22, 68-71 (2009).
Jiang, S. & Dong, C. A Complex issue on CD4 ' T-cell subsets. Immunol Rev.
252, 5-11
(2013).
June, C.H. et at. Inhibition of tyrosine phosphorylation prevents T-cell
receptor-mediated
signal transduction. Proc.Natl.Acad.Sci. 87, 7722-7726 (1990).
Kapturczak, M.H. et at. Pharmacology of calcineurin antagonists. Transplant
Proc. 36, 25-
32 (2004).
Kelly, W.D. et at. Allotransplantation of the pancreas and duodenum along with
the kidney
in diabetic nephropathy. Surgery 61, 827-837 (1967).
Landsteiner, K. Ueber Agglutinationserscheinungen normalen menschlichen
Blutes.
Wiener Klinische Wochenschrift 49, 1132-1133 (1901).
Laub R. et at. A multiple transgenic mouse model with a partially humanized
activation
pathway for helper T cell responses. J. Immunol. Methods 246, 37-50 (2000).
Laub, R., Dorsch, M., Wenk, K., & Emmrich, F. Induction of immunologic
tolerance to
tetanus toxoid by anti-human CD4 in HLA-DR3(+)/human CD4(+)/murine CD4(-)
multiple transgenic mice. Transplant. Proc. 33, 2182-2183 (2001).
Laub, R. et at. Anti-human CD4 induces peripheral tolerance in a human CD4+,
murine
CD4-, HLA-DR+ advanced transgenic mouse model. J. lmmunol. 169, 2947-2955
(2002).

CA 03012277 2018-07-23
WO 2017/140735 - 41 - PCT/EP2017/053417
Liu,W. et at. CD127 expression inversely correlates with FoxP3 and suppressive
function
of human CD4+ T reg cells. J. Exp. Med. 203, 1701-1711 (2006).
Luckheeram, R.V. et at. CD4 ' T cells: differentiation and functions. Clin Dev
Immunol.
2012, 1-12 (2012).
Madrenas, J., Schwartz, R.H., & Germain,R.N. Interleukin 2 production, not the
pattern of
early T-cell antigen receptor-dependent tyrosine phosphorylation, controls
anergy
induction by both agonists and partial agonists. Proc. Nat!. Acad. Sci. U. S.
A 93, 9736-
9741 (1996).
Madrenas, J. & Germain, R.N. Variant TOR ligands: new insights into the
molecular basis
of antigen-dependent signal transduction and T-cell activation. Semin.
Immunol. 8, 83- 101
(1996).
Medawar, P.B. The behaviour and fate of skin autografts and skin homografts in
rabbits. J
Anat. 78, 176-199 (1944).
Murray, J.E. et at. Renal transplantation: a twenty-five year experience. Ann
Surg. 184,
565-573 (1976).
O'Connell, R.M. et at. MicroRNA-155 promotes autoimmune inflammation by
enhancing
inflammatory T cell development. Immunity. 33, 607-619 (2010).
Pfitzmann R, Neuhau P, Hetzer R (Hrsg.): Organtransplantation, ISBN 3-11-
016849-9
Reinke, P. et at. Anti-CD4 therapy of acute rejection in long-term renal
allograft recipients.
Lancet 338. 702-703 (1991).
Reinke, P. et at. Late acute rejection in long-term renal allograft
recipients. Diagnostic and
predictive value of circulating activated T cells. Transplantation 58, 35-41
(1994).
Reinke, P. et at. Anti-CD4 monoclonal antibody therapy of late acute rejection
in renal
allograft recipients--CD4+ T cells play an essential role in the rejection
process.
Transplant. Proc. 27, 859-862 (1995).
Robey, E. & Axel, R. CD4:Collaborator in immune recognition and HIV infection.
Cell
60, 697-700 (1990).
Schmidt F. et at., Flow cytometric analysis of the graft-versus-Leukemia-
effect after
hematopoietic stem cell transplantation in mice. Cytometry A. 87(4): 334-45
(2015).
Stewart, S.J. et at. Human T lymphocytes and monocytes bear the same Leu-3(T4)
antigen.
J Immunol. 136, 3773-3778 (1986).
Sayegh, M.H. et at. Transplantation 50 Years Later-Progress, Challenges, and
Promises. N
Engl J Med. 351, 2761-2766 (2004).
Schulz, K.-H. & Koch, U. Transplantationspsychiologie. In Balck, F.
Anwendungsfelder
der medizinischen Psychiologie. Springer, 101-114 (2005).
Starzl, T.E. et at. Orthotopic homotransplantation of the human liver. Ann
Surg. 168, 392-
415 (1968).
Starzl, T.E. et at. Evolution of liver transplantation. Hepatology 2, 614-636
(1982).
Strauss, G., Vignali.D.A.. Schonrich,G.. & Hammerling,G.J. Negative and
positive
selection by HLA-DR3(DRw17) molecules in transgenic mice. Immunogenetics 40.
104-
108 (1994).

CA 03012277 2018-07-23
WO 2017/140735 PCT/EP2017/053417
42
Applicant's or agent's International application No.
file reference F68779pc PCT/EP2017/053417
INDICATIONS RELATING TO DEPOSITED MICROORGANISM
OR OTHER BIOLOGICAL MATERIAL
(PCT Rule 13 bis)
A. The indications made below relate to the deposited microorganism or other
biological material referred to in the description
on page 31 , line 7
B. IDENTIFICATION OF DEPOSIT Further deposits are identified on an
additional sheet E
Name of depositary institution
ECACC European Collection of Cell Cultures (ECACC) Culture Collections Public
Health England
Address of depositary institution (including postal code and country)
Porton Down
Salisbury, Wiltshire SP4 OJG
United Kingdom
Date of deposit Accession Number
05/05/1988 88050502
C. ADDITIONAL INDICATIONS (leave blank if not applicable) This information
is continued on an additional sheet
D. DESIGNATED STATES FOR WHICH INDICATIONS ARE MADE (([the indications are not
for all designated States)
E. SEPARATE FURNISHING OF INDICATIONS (leave blank ([not applicable)
The indications listed below will be submitted to the International Bureau
later (spec()5)the general nature of the indic,ations e.g, "Accession
Number of Deposit')
______________________________________________ For receiving Office use only
For International Bureau use only
This sheet was received with the international application 0 This sheet was
received by the International Bureau on:
Authorized officer Authorized officer
Kuiper¨Cristina, Nathalie
Form PCT/R0/134 (July1998; reprint January 2004)
SUBSTITUTE SHEET (RULE 26)

CA 03012277 2018-07-23
WO 2017/140735 PCT/EP2017/053417
43
Applicant's or agent's International application No.
file reference F68779pc PCT/EP2017/053417
INDICATIONS RELATING TO DEPOSITED MICROORGANISM
OR OTHER BIOLOGICAL MATERIAL
(PCT Rule 13bis)
A. The indications made below relate to the deposited microorganism or other
biological material referred to in the description
on page 31 ,line 27
B. IDENTIFICATION OF DEPOSIT Further deposits are identified on an
additional sheet K
Name of depositary institution
DSMZ Leibniz-lnstitut DSMZ - Deutsche Sammlung von Mikroorganismen und Zell-
kulturen GmbH (DSMZ)
Address of depositary institution (including postal code and country)
Inhoffenstr. 7B
38124 Braunschweig
Germany
Date of deposit Accession Number
02/12/2011 DSM ACC3148
C. ADDITIONAL INDICATIONS (leave blank if not applicable) This information
is continued on an additional sheet
D. DESIGNATED STATES FOR WHICH INDICATIONS ARE MADE (if the indications are
not for all designated States)
E. SEPARATE FURNISHING OF INDICATIONS (leave blank if not applicable)
The indications listed below will be submitted to the International Bureau
later (specifr the general nature of the indications e.g., "Accession
Number of Deposit")
______________________________________________ For receiving Office use only
For International Bureau use only
This sheet was received with the international application Eli This sheet
was received by the International Bureau on:
Authorized officer Authorized officer
Kuiper¨Cristina, Nathalie
Form PCT/RO/134 (July1998; reprint January 2004)
SUBSTITUTE SHEET (RULE 26)

CA 03012277 2018-07-23
WO 2017/140735 PCT/EP2017/053417
44
Applicant's or agent's International application No.
file reference F68779pc PCT/EP2017/053417
INDICATIONS RELATING TO DEPOSITED MICROORGANISM
OR OTHER BIOLOGICAL MATERIAL
(PCT Rule 13bis)
A. The indications made below relate to the deposited microorganism or other
biological material referred to in the description
on page 31 , line 32
B. IDENTIFICATION OF DEPOSIT Further deposits are identified on an
additional sheet El
Name of depositary institution
DSMZ Leibniz-lnstitut DSMZ - Deutsche Sammlung von Mikroorganismen und Zell-
kulturen GmbH (DSMZ)
Address of depositary institution (including postal code and country)
Inhoffenstr. 7B
38124 Braunschweig
Germany
Date of deposit Accession Number
02/12/2011 DSM ACC3147
C. ADDITIONAL INDICATIONS (leave blank if not applicable) This information
is continued on an additional sheet
D. DESIGNATED STATES FOR WHICH INDICATIONS ARE MADE (f the indications are not
for all designated Stales)
E. SEPARATE FURNISHING OF INDICATIONS (leave blank i f not applicable)
The indications listed below will be submitted to the International Bureau
later (speco, the general nature of the indications e.g., 'Accession
Number ofDeposit")
______________________________________________ For receiving Office use only
For International Bureau usc only
This sheet was received with the international application This sheet was
received by the International Bureau on:
Authorized officer Authorized officer
Kuiper¨Cristina, Nathalie
Form PCT/R0/134 (July1998; reprint January 2004)
SUBSTITUTE SHEET (RULE 26)

CA 03012277 2018-07-23
WO 2017/140735
PCT/EP2017/053417
% ECACC
5,.L:perxr
Vet-1C:
NIX="ir=V'VP-sr=Vr'urn3
EtropeanCeectcn O Ce C1ures
Heat), Pootecucen Agency
Cenue lor Emergency Pepa-edress ard Pest-m.1R
Ptrtosi Crcnvn, Sa'sbry SF4 OtG undud Klagdrim
=-4.4 tO MC 611512 0., +43 C11920611315
npacat; pa g wow Pei tru,
Health Protection Agency
Porton Down
And
European Collection of Cell Cultures
This document certifies that:
Cell Line 30E16H5 (New stock received 2012 for a further 30 years)
Deposit Reference 88050502
Has been accepted as a patent deposit, in accordance with
The Budapest Treaty of 1977
With the European Collection of Cell Cultures on
05 May 1988
ECACC Patent Supervisor

CA 03012277 2018-07-23
WO 2017/140735 PCT/EP2017/053417
46
.1/4., IflL LSUS.jjl I ...Jr
FOR THE PURPOSES OF PATENT PROCED LIRE
INTERNA TIONAL 'FORM
Fraunhofer Gesellschaft zur Forderung
der angewandlen Forschung e,V,
Hansastr. 27c RECEPT IN THE CASE OF AN ctranINAL
'Dz.posrr
issued pursuant to Rule 73 by the
S0686 Munchen INTERNATIONAL DEPOSITARY AUTHORITY
ident ifie d at the bottom or this paga
L IDENTIFICATION Of THE MICROORGANISM
Ithmtifiestion refers-ins given by the DEPOSITOR: Accessional/mbar given by
the
MAX.161-15/30F16H5 acrtINATIONAL DEPOSITARY AUTHORITY:
11)SM ACC314/3
IL SCIENIIHC DESCRIPTION AND/OR PROPOSED TAXONOMIC DESIGNATION
The microorganism identified under Low so avcompujitd by-
x t0fentinV description
I I a proposed taxonomic designation
Nark 1LS a cp0s4 slitsC7t applicable).
HI. RECEIPT AND ACCEPTANCE
=
This Imernational Depositary Authority accepts the microorganism identified
uncle.; I. above, whial was received by It on 2011-12-02
(Dale oldie original deporit)'.
IV, RECEIPT OF REQUEST FOR roNVERSION
The microorganism ides:tined under shiwe was received by this bgemntionsl
Depositary Authority on (date of origsral deposi0
and a request in COTWCLI dlt original deposit to a deposit under she Budapest
Treaty was receiva by it -on (dare of receip; of request
for conversion).
V. INTERNATIONAL DEPOSITARY AtrnioRrre
Name: DS:MZ-DEUTSCIIE SAMMLUNO VON Si gnaturc(s) of persorlIsI having
the power to reprment the
MIKROORGANISMEN UND ZELLKIJLTUREN GmbH Internatioruil Depositary Authanty
co of authorized official(s):
Address: Inhoirensir. 7 B
0-311 124 Braunschweig
. . .
. =
Date: 20.11- 12-16
' Where Rule 6,4 (d) applies, such data is the dam on which the status of
international dcposiutriauthority was acquired.
Form DSMZ-UP/4 (sok page) O812006

CA 03012277 2018-07-23
WO 2017/140735
PCT/EP2017/053417
47
BUDAPEST TREATY ON THE INTERNATIONAL
RECOGNITION or THE DEPOSIT OF MICROORGANISMS
FOR TIIE PURPOSES OF PATENT PROCEDURE
Ice.
INTERNATIONAL FORM
Fraunhofer-GeselIschaft zur Forderung
der angewandten Forschung
Flansastr, 27c RECEIPT IN THE CASE OF AN ORIGINAL
DEPOSIT
issued pursuant To Rule 7.1 by am
80686 Munchen INTERNATIONAL DEPOSITARY AUTHORITY
identified at the bonom of this page
1_ IDENTIFICATION OF THE MICROORGANISM
Identification reference given by the DEPOSITOR: Accession amber given by
the
LNTERNATIONAL DEPOSITARY AUTHORITY:
CD4.161-15.chimleG-1
DSM ACC3147
II. SCIENTIFIC DESCRUPTION AND/OR PROPOSED TAXONOMIC DESIGNATION
The microorganism identified under I. above svas accomporned by:
) soicritiric description
( I a proposed Taxonomic designation
CMark with a cross where applicab1c).
ID. RECEIPT AND ACCEPTANCE
This International Depositary Authority accepts the microotganinn klened
aruier 1. above. which was received by it an 2011-12-02
(Dote of the original deposit/I.
IV. RECEIPT OF REQUEST FOR CONVERSION
The onkroorga Mena ideetified under I above twat received by this knernaaanal
Depositary Authority an fdatr of original deposit)
and o request to conven The original deposit To a deposit undll the Budapest
Treaty was rteeived by it on telatc of receipt of rcquest
Or conversion).
V. INTERNATIONAL DEPOSITARY mmi0g1TY
Name: EISMZ-DEUTSCHE SANIM LUNG VON Signature(s) of person{s) having the
pots-cr to rt-perseal the
MIK ROORGANfISM EN LIND ZELLKULTUREN GtnH
ernational Depositary Authority ce af outhOrt2ed frit IWO:
Address: 401oltensor. 7
D-38124 Braunschweig 6/ 4,)'0.----Z4
Dale: 2011-12-16
' wrwr, R.ttle 6,4 (d) applies, such ctssr is the dote no which the
artatias cf i.trrnEciion3i depositary autaarily was aaar4ress.
Form PS b1Z-13F14 (sole page) 02.17006

Representative Drawing

Sorry, the representative drawing for patent document number 3012277 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2017-02-15
(87) PCT Publication Date 2017-08-24
(85) National Entry 2018-07-23
Examination Requested 2021-10-28

Abandonment History

There is no abandonment history.

Maintenance Fee

Last Payment of $277.00 was received on 2024-01-29


 Upcoming maintenance fee amounts

Description Date Amount
Next Payment if small entity fee 2025-02-17 $100.00
Next Payment if standard fee 2025-02-17 $277.00 if received in 2024
$289.19 if received in 2025

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2018-07-23
Maintenance Fee - Application - New Act 2 2019-02-15 $100.00 2018-07-23
Maintenance Fee - Application - New Act 3 2020-02-17 $100.00 2020-01-30
Maintenance Fee - Application - New Act 4 2021-02-15 $100.00 2021-02-10
Request for Examination 2022-02-15 $816.00 2021-10-28
Maintenance Fee - Application - New Act 5 2022-02-15 $203.59 2022-02-08
Maintenance Fee - Application - New Act 6 2023-02-15 $210.51 2023-02-06
Maintenance Fee - Application - New Act 7 2024-02-15 $277.00 2024-01-29
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
FRAUNHOFER-GESELLSCHAFT ZUR FOERDERUNG DER ANGEWANDTEN FORSCHUNG E.V.
Past Owners on Record
None
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Request for Examination 2021-10-28 4 167
Examiner Requisition 2022-11-23 7 441
Amendment 2023-03-21 116 6,818
Description 2023-03-21 41 3,476
Claims 2023-03-21 6 277
Abstract 2018-07-23 1 59
Claims 2018-07-23 6 247
Drawings 2018-07-23 3 246
Description 2018-07-23 47 2,735
Patent Cooperation Treaty (PCT) 2018-07-23 1 55
International Search Report 2018-07-23 3 96
National Entry Request 2018-07-23 7 209
Cover Page 2018-08-02 1 35
Examiner Requisition 2024-06-13 7 459

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :